(19)
(11) EP 2 468 770 B1

(12) EUROPEAN PATENT SPECIFICATION

(45) Mention of the grant of the patent:
20.12.2017 Bulletin 2017/51

(21) Application number: 11192705.9

(22) Date of filing: 13.07.2007
(51) International Patent Classification (IPC): 
C07K 16/18(2006.01)
A61P 25/28(2006.01)

(54)

Humanized antibody against amyloid beta.

Humanisierte Antikörper gegen Amyloid beta.

Anticorps humanisés contre beta amyloide.


(84) Designated Contracting States:
AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC MT NL PL PT RO SE SI SK TR
Designated Extension States:
RS

(30) Priority: 11.06.2007 US 94328907 P
12.06.2007 US 94349907 P
14.07.2006 EP 06014730
02.10.2006 EP 06020765

(43) Date of publication of application:
27.06.2012 Bulletin 2012/26

(60) Divisional application:
17190333.9

(62) Application number of the earlier application in accordance with Art. 76 EPC:
07840408.4 / 2046833

(73) Proprietors:
  • AC Immune S.A.
    1015 Lausanne (CH)
  • Genentech, Inc.
    South San Francisco, CA 94080-4990 (US)

(72) Inventors:
  • Pfeifer, Andrea
    1806 St.-Légier (CH)
  • Pihlgren Bosch, Maria
    1052 Mont-sur-Lausanne (CH)
  • Muhs, Andreas
    1053 Cugny (CH)
  • Watts, Ryan
    San Mateo, CA 94402 (US)

(74) Representative: Weber, Martin et al
Jones Day Prinzregentenstraße 11
80538 München
80538 München (DE)


(56) References cited: : 
WO-A-00/72880
WO-A-02/096937
WO-A-2006/055178
WO-A-2007/068412
WO-A-01/62801
WO-A-03/070760
WO-A-2006/066171
   
  • BARD F ET AL: "Epitope and isotype specificities of antobodies to beta-amyloid peptide for protection against Alzheimer's disease-like neuropathology", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE, WASHINGTON, DC, US, vol. 100, no. 4, 18 February 2003 (2003-02-18), pages 2023-2028, XP002982464, ISSN: 0027-8424
  • LIU RUITIAN ET AL: "Single chain variable fragments against beta-amyloid (Abeta) can inhibit Abeta aggregation and prevent Abeta-induced neurotoxicity", BIOCHEMISTRY, AMERICAN CHEMICAL SOCIETY. EASTON, PA, US, vol. 43, no. 22, 12 May 2004 (2004-05-12), pages 6959-6967, XP002395890, ISSN: 0006-2960
  • SOLOMON B ET AL: "MONOCLONAL ANTIBODIES INHIBIT IN VITRO FIBRILLAR AGGREGATION OF THE ALZHEIMER BETA-AMYLOID PEPTIDE", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE, WASHINGTON, DC, US, vol. 93, January 1996 (1996-01), pages 452-455, XP002942850, ISSN: 0027-8424
  • RUDIKOFF S ET AL: "Single amino acid substitution altering antigen-binding specificity", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, NATIONAL ACADEMY OF SCIENCES, US, vol. 79, 1 March 1982 (1982-03-01), pages 1979-1983, XP007901436, ISSN: 0027-8424, DOI: 10.1073/PNAS.79.6.1979
   
Note: Within nine months from the publication of the mention of the grant of the European patent, any person may give notice to the European Patent Office of opposition to the European patent granted. Notice of opposition shall be filed in a written reasoned statement. It shall not be deemed to have been filed until the opposition fee has been paid. (Art. 99(1) European Patent Convention).


Description


[0001] The present invention is related to antibodies and compositions for diagnosis and treatment of amyloidosis, a group of disorders and abnormalities associated with amyloid protein such as Alzheimer's disease.

[0002] Amyloidosis is not a single disease entity but rather a diverse group of progressive disease processes characterized by extracellular tissue deposits of a waxy, starch-like protein called amyloid, which accumulates in one or more organs or body systems. As the amyloid deposits accumulate, they begin to interfere with the normal function of the organ or body system. There are at least 15 different types of amyloidosis. The major forms are primary amyloidosis without known antecedent, secondary amyloidosis following some other condition, and hereditary amyloidosis.

[0003] Secondary amyloidosis occurs during chronic infection or inflammatory disease, such as tuberculosis, a bacterial infection called familial Mediterranean fever, bone infections (osteomyelitis), rheumatoid arthritis, inflammation of the small intestine (granulomatous ileitis), Hodgkin's disease, and leprosy.

[0004] Amyloid deposits include amyloid P (pentagonal) component (AP), a glycoprotein related to normal serum amyloid P (SAP), and sulphated glycosaminoglycans (GAG), complex carbohydrates of connective tissue. Amyloid protein fibrils, which account for about 90% of the amyloid material, comprise one of several different types of proteins. These proteins are capable of folding into so-called "beta-pleated" sheet fibrils, a unique protein configuration which exhibits binding sites for Congo red resulting in the unique staining properties of the amyloid protein.

[0005] Many diseases of aging are based on or associated with amyloid-like proteins and are characterized, in part, by the buildup of extracellular deposits of amyloid or amyloid-like material that contribute to the pathogenesis, as well as the progression of the disease. These diseases include, but are not limited to, neurological disorders such as Alzheimer's Disease (AD), Lewy body dementia, Down's syndrome, hereditary cerebral hemorrhage with amyloidosis (Dutch type); the Guam Parkinson-Dementia complex. Other diseases which are based on or associated with amyloid-like proteins are progressive supranuclear palsy, multiple sclerosis; Creutzfeld Jacob disease, Parkinson's disease, HIV-related dementia, ALS (amyotropic lateral sclerosis), Adult Onset Diabetes; senile cardiac amyloidosis; endocrine tumors, and others, including macular degeneration.

[0006] Although pathogenesis of these diseases may be diverse, their characteristic deposits often contain many shared molecular constituents. To a significant degree, this may be attributable to the local activation of pro-inflammatory pathways thereby leading to the concurrent deposition of activated complement components, acute phase reactants, immune modulators, and other inflammatory mediators (McGeer et al., 1994).

[0007] Alzheimer's Disease (AD) is a neurological disorder primarily thought to be caused by amyloid plaques, an accumulation of abnormal deposit of proteins in the brain. The most frequent type of amyloid found in the brain of affected individuals is composed primarily of Aβ fibrils. Scientific evidence demonstrates that an increase in the production and accumulation of beta-amyloid protein in plaques leads to nerve cell death, which contributes to the development and progression of AD. Loss of nerve cells in strategic brain areas, in turn, causes reduction in the neurotransmitters and impairment of memory. The proteins principally responsible for the plaque build up include amyloid precursor protein (APP) and two presenilins (presenilin I and presenilin II). Sequential cleavage of the amyloid precursor protein (APP), which is constitutively expressed and catabolized in most cells, by the enzymes β and γ secretase leads to the release of a 39 to 43 amino acid Aβ peptide. The degradation of APPs likely increases their propensity to aggregate in plaques. It is especially the Aβ(1-42) fragment that has a high propensity of building aggregates due to two very hydrophobic amino acid residues at its C-terminus. The Aβ(1-42) fragment is therefore believed to be mainly involved and responsible for the initiation of neuritic plaque formation in AD and to have, therefore, a high pathological potential. There is therefore a need for agents to prevent the formation of amyloid plaques and to diffuse existing plaques in AD.

[0008] The symptoms of AD manifest slowly and the first symptom may only be mild forgetfulness. In this stage, individuals may forget recent events, activities, the names of familiar people or things and may not be able to solve simple math problems. As the disease progresses, symptoms are more easily noticed and become serious enough to cause people with AD or their family members to seek medical help. Mid-stage symptoms of AD include forgetting how to do simple tasks such as grooming, and problems develop with speaking, understanding, reading, or writing. Later stage AD patients may become anxious or aggressive, may wander away from home and ultimately need total care.

[0009] Presently, the only definite way to diagnose AD is to identify plaques and tangles in brain tissue in an autopsy after death of the individual. Therefore, doctors can only make a diagnosis of "possible" or "probable" AD while the person is still alive. Using current methods, physicians can diagnose AD correctly up to 90 percent of the time using several tools to diagnose "probable" AD. Physicians ask questions about the person's general health, past medical problems, and the history of any difficulties the person has carrying out daily activities. Behavioral tests of memory, problem solving, attention, counting, and language provide information on cognitive degeneration and medical tests such as tests of blood, urine, or spinal fluid, and brain scans can provide some further information.

[0010] The management of AD consists of medication-based and non-medication based treatments. Treatments aimed at changing the underlying course of the disease (delaying or reversing the progression) have so far been largely unsuccessful. Medicines that restore the deficit (defect), or malfunctioning, in the chemical messengers of the nerve cells (neurotransmitters), in particular the cholinesterase inhibitors (ChEIs) such as tacrine and rivastigmine, have been shown to improve symptoms. ChEIs impede the enzymatic degradation of neurotransmitters thereby increasing the amount of chemical messengers available to transmit the nerve signals in the brain.

[0011] For some people in the early and middle stages of the disease, the drugs tacrine (COGNEX®, Morris Plains, NJ), donepezil (ARICEPT®, Tokyo, JP), rivastigmine (EXELON®, East Hanover, NJ), or galantamine (REMINYL®, New Brunswick, NJ) may help prevent some symptoms from becoming worse for a limited time. Another drug, memantine (NAMENDA®, New York, NY), has been approved for treatment of moderate to severe AD. Medications are also available to address the psychiatric manifestations of AD. Also, some medicines may help control behavioral symptoms of AD such as sleeplessness, agitation, wandering, anxiety, and depression. Treating these symptoms often makes patients more comfortable and makes their care easier for caregivers. Unfortunately, despite significant treatment advances showing that this class of agents is consistently better than a placebo, the disease continues to progress, and the average effect on mental functioning has only been modest. Many of the drugs used in AD medication such as, for example, ChEIs also have side effects that include gastrointestinal dysfunction, liver toxicity and weight loss.

[0012] Another disease that is based on or associated with the accumulation and deposit of amyloid-like protein is macular degeneration.

[0013] Macular degeneration is a common eye disease that causes deterioration of the macula, which is the central area of the retina (the paper-thin tissue at the back of the eye where light-sensitive cells send visual signals to the brain). Sharp, clear, 'straight ahead' vision is processed by the macula. Damage to the macula results in the development of blind spots and blurred or distorted vision. Age-related macular degeneration (AMD) is a major cause of visual impairment in the United States and for people over age 65 it is the leading cause of legal blindness among Caucasians. Approximately 1.8 million Americans age 40 and older have advanced AMD, and another 7.3 million people with intermediate AMD are at substantial risk for vision loss. The government estimates that by 2020 there will be 2.9 million people with advanced AMD. Victims of AMD are often surprised and frustrated to find out how little is known about the causes and treatment of this blinding condition.

[0014] There are two forms of macular degeneration: dry macular degeneration and wet macular degeneration. The dry form, in which the cells of the macula slowly begin to break down, is diagnosed in 85 percent of macular degeneration cases. Both eyes are usually affected by dry AMD, although one eye can lose vision while the other eye remains unaffected. Drusen, which are yellow deposits under the retina, are common early signs of dry AMD. The risk of developing advanced dry AMD or wet AMD increases as the number or size of the drusen increases. It is possible for dry AMD to advance and cause loss of vision without turning into the wet form of the disease; however, it is also possible for early-stage dry AMD to suddenly change into the wet form.
The wet form, although it only accounts for 15 percent of the cases, results in 90 percent of the blindness, and is considered advanced AMD (there is no early or intermediate stage of wet AMD). Wet AMD is always preceded by the dry form of the disease. As the dry form worsens, some people begin to have abnormal blood vessels growing behind the macula. These vessels are very fragile and will leak fluid and blood (hence 'wet' macular degeneration), causing rapid damage to the macula.

[0015] The dry form of AMD will initially often cause slightly blurred vision. The center of vision in particular may then become blurred and this region grows larger as the disease progresses. No symptoms may be noticed if only one eye is affected. In wet AMD, straight lines may appear wavy and central vision loss can occur rapidly.

[0016] Diagnosis of macular degeneration typically involves a dilated eye exam, visual acuity test, and a viewing of the back of the eye using a procedure called fundoscopy to help diagnose AMD, and-if wet AMD is suspected-fluorescein angiography may also be performed. If dry AMD reaches the advanced stages, there is no current treatment to prevent vision loss. However, a specific high dose formula of antioxidants and zinc may delay or prevent intermediate AMD from progressing to the advanced stage. Macugen® (pegaptanib sodium injection), laser photocoagulation and photodynamic therapy can control the abnormal blood vessel growth and bleeding in the macula, which is helpful for some people who have wet AMD; however, vision that is already lost will not be restored by these techniques. If vision is already lost, low vision aids exist that can help improve the quality of life.

[0017] One of the earliest signs of age-related macular degeneration (AMD) is the accumulation of extracellular deposits known as drusen between the basal lamina of the retinal pigmented epithelium (RPE) and Bruch's membrane (BM). Recent studies conducted by Anderson et al. have confirmed that drusen contains amyloid beta. (Experimental Eye Research 78 (2004) 243-256).

[0018] Ongoing research continues with studies exploring environmental, genetic, and dietary factors that may contribute to AMD. New treatment strategies are also being explored, including retinal cell transplants, drugs that will prevent or slow down the progress of the disease, radiation therapy, gene therapies, a computer chip implanted in the retina that may help stimulate vision and agents that will prevent the growth of new blood vessels under the macula.

[0019] An important factor to consider when developing new drugs is the ease of use for the target patients. Oral drug delivery, -specifically tablets, capsules and softgels-, account for 70% of all dosage forms consumed because of patient convenience. Drug developers agree that patients prefer oral delivery rather than subjecting themselves to injections or other, more invasive forms of medicinal administration. Formulations resulting in low dosing intervals (i.e. once a day or sustained release) are also preferable. The ease of administering antibiotics in oral dosage forms results in an increase of patient compliance during treatment.

[0020] What is needed are effective methods and compositions for preventing or addressing the complications associated with amyloidosis, a group of diseases and disorders associated with amyloid plaque formation including secondary amyloidosis and age-related amyloidosis including, but not limited to, neurological disorders such as Alzheimer's Disease (AD), Lewy body dementia, Down's syndrome, hereditary cerebral hemorrhage with amyloidosis (Dutch type); the Guam Parkinson-Dementia complex; as well as other diseases which are based on or associated with amyloid-like proteins such as progressive supranuclear palsy, multiple sclerosis; Creutzfeld Jacob disease, Parkinson's disease, HIV-related dementia, ALS (amyotropic lateral sclerosis), Adult Onset Diabetes; senile cardiac amyloidosis; endocrine tumors, and others, including macular degeneration. In particular what is needed are agents capable of counteracting the physiological manifestations of the disease such as the formation of plaques associated with aggregation of fibers of the amyloid or amyloid-like peptide.

[0021] Anti-amyloid antibodies elicited by the inoculation of Aβ1-42 mixed with Freund complete or incomplete adjuvant were reported to reduce the amyloid burden in transgenic mice for human Alzheimer disease (Schenk et al., 1999). Intraperitoneal inoculation of tetrapalmitoylated Aβ1-16 reconstituted in liposomes to NORBA transgenic mice elicited significant titers of anti-amyloid antibodies, which were reported to solubilize amyloid fibers and plaques in vitro and in vivo. (Nicolau et al., 2002).

[0022] WO 01/62801 A2 discloses an anti-β-amyloid humanized antibody of the 266 antibody and suggests that antibodies that bind β-amyloid between positions 13 and 28 (e.g., 266 and 4G8) are capable of sequestering soluble forms of β-amyloid from their bound, circulating forms in the blood without binding with great affinity to aggregated β-amyloid.

[0023] WO 03/070760 A2 discloses an antibody molecule that recognizes a conformational epitope on the β-A4 peptide, which is defined by the amino acid sequences AEFRHDSGY and VHHQKLVFFAEDVG.

[0024] WO 2006/066171 A1 discloses an anti-13-amyloid antibody (15C11) that binds to the central region of β-amyloid (i.e., amino acids 19-22) and binds oligomeric β-amyloid species, but does not appear to bind to β-amyloid monomers.

[0025] A possible mechanism by which the dissolution of amyloid plaques and fibres occurred was first suggested by Bard et al., (2000), who concluded that the antibodies opsonized the plaques, which were subsequently destroyed by the macrophages of the microglia. De Mattos et al., (2001) indicated that a mAb directed against the central domain of β-amyloid was able to bind and completely sequester plasma amyloid. They argued that the presence of these mAbs in circulation shifted the equilibrium of Aβ between brain and plasma, favoring the peripheral clearing and catabolism instead of deposition within the brain.

[0026] Prolonged human therapy with rodent antibodies may result in an antiglobulin response which is detectable at about 8-12 days after administration and reaches a peak at about 20-30 days. If such an antiglobulin response is encountered, the treatment must be discontinued after not more than about 10 days and re-treatment at a latter date is usually precluded because it will lead to rapid onset of a secondary antiglobulin response. Although rodent antibodies share a considerable degree of sequence conservation with that of human antibodies, there are many sequence differences between rodents and human antibodies sufficient for the rodent antibodies to be immunogenic in humans.

[0027] This problem may be overcome by generating antibodies directly in humans or by the creation of "humanized' (a.k.a. "reshaped' antibodies). Humanized antibodies have a variable region amino acid sequence that contains the rodent-derived CDRs interspersed into human or human-like framework sequences. Since the specificity of the humanized antibody is provided by the rodent-derived CDRs, their residues are to be used essentially unchanged with only minor modifications being allowable, which do not significantly interfere with the affinity and specificity of the antibody for its target antigen. Framework residues may be derived from any primate or, particularly, from any human variable region or may be a combination thereof and the resultant designed variable region would be considered reshaped.

[0028] To maximise the likelihood that affinity will be retained in the reshaped antibody it is important to make a proper selection of the framework region. It is known that the framework sequences serve to hold the CDRs in their correct spatial orientation for interaction with antigen, and that framework residues can sometimes even participate in antigen binding. In order to maintain the affinity of the antibody for its antigen it is advantageous to select human framework sequences that are most similar to the sequences of the rodent frameworks. It then may still be necessary to replace one or more amino acids in the human framework sequence with the corresponding residue in the rodent framework to avoid losses with the affinity. This replacement may be aided by computer modelling.

[0029] The present invention provides novel antibodies and compositions comprising highly specific and highly effective antibodies, namely humanized antibodies including fragments thereof, having the ability to specifically recognize and bind to specific epitopes from a range of β-amyloid antigens, which my be presented to the antibody in a monomeric, dimeric, trimeric, etc, a polymeric form, in form of an aggregate, fibers, filaments or in the condensed form of a plaque. The antibodies enabled by the teaching of the present invention are particularly useful for the treatment of amyloidosis, a group of diseases and disorders associated with amyloid plaque formation including secondary amyloidosis and age-related amyloidosis including, but not limited to, neurological disorders such as Alzheimer's Disease (AD), Lewy body dementia, Down's syndrome, hereditary cerebral hemorrhage with amyloidosis (Dutch type); the Guam Parkinson-Dementia complex; as well as other diseases which are based on or associated with amyloid-like proteins such as progressive supranuclear palsy, multiple sclerosis; Creutzfeld Jacob disease, hereditary cerebral hemorrhage with amyloidosis Dutch type, Parkinson's disease, HIV-related dementia, ALS (amyotropic lateral sclerosis), Adult Onset Diabetes; senile cardiac amyloidosis; endocrine tumors, and others, including macular degeneration, to name just a few.

[0030] Surprisingly, the antibodies of the invention bind to β-amyloid in various forms of β-amyloid, including soluble monomers, oligomers and fibrils, with high affinity, and they inhibit aggregation of monomers into oligomers/fibrils and induce disaggregation of oligomers/fibrils.

[0031] The invention provides a humanized antibody or a fragment thereof, wherein said antibody or fragment thereof comprises:
  1. (A) (i) a Heavy Chain Variable Region (HCVR) having an amino acid sequence that is 96%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 15, wherein the HCVR comprises a heavy chain complementarity determining region 1 (CDR1) as given in SEQ ID NO: 1, a heavy chain CDR2 as given in SEQ ID NO: 2 and a heavy chain CDR3 as given in SEQ ID NO: 3, and (ii) a light chain variable region (LCVR) comprising a light chain CDR1 as given in SEQ ID NO: 4, a light chain CDR2 as given in SEQ ID NO: 5, RVSNRFS or KVSSRFS and a light chain CDR3 as given in SEQ ID NO: 6, or
  2. (B) (i) an HCVR comprising a heavy chain CDR1 as given in SEQ ID NO: 1, a heavy chain CDR2 as given in SEQ ID NO: 2 and a heavy chain CDR3 as given in SEQ ID NO: 3, and (ii) a LCVR having an amino acid sequence that is 96%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 12, wherein the LCVR comprises a light chain CDR1 as given in SEQ ID NO: 4, a light chain CDR2 as given in SEQ ID NO: 5, RVSNRFS or KVSSRFS and a light chain CDR3 as given in SEQ ID NO: 6,
wherein said antibody or fragment thereof specifically binds to β-amyloid protein.

[0032] In some embodiments, the humanized antibody of the invention or fragment thereof as defined in 1(A) above, comprises the LCVR having an amino acid sequence that is 96%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 12.

[0033] In some embodiments, the humanized antibody of the invention or fragment thereof as defined in 1(B) above, comprises the HCVR having an amino acid sequence that is 96%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 15.

[0034] In some embodiments, the humanized antibody of the invention or a fragment thereof comprises:
  1. i) the HCVR having the amino acid sequence of SEQ ID NO: 15 or an amino acid sequence that is 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 15; or
  2. ii) the LCVR having the amino acid sequence of SEQ ID NO: 12 or an amino acid sequence that is 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 12; or
  3. iii) the HCVR having the amino acid sequence of SEQ ID NO: 15 or an amino acid sequence that is 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 15 and the LCVR having the amino acid sequence of SEQ ID NO: 12 or an amino acid sequence that is 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 12.


[0035] In some embodiments, the humanized antibody of the invention or fragment thereof comprises:
  1. i) a heavy chain comprising the amino acid sequence of SEQ ID NO: 16 or an amino acid sequence that is 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 16; or
  2. ii) a light chain comprises the amino acid sequence of SEQ ID NO: 13 or an amino acid sequence that is 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 13; or
  3. iii) a heavy chain comprising the amino acid sequence of SEQ ID NO: 16 or an amino acid sequence that is 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 16 and a light chain comprising the amino acid sequence of SEQ ID NO: 13 or an amino acid sequence that is 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 13.


[0036] In some embodiments, the humanized antibody of the invention or fragment thereof is of the IgG4 isotype.

[0037] In some embodiments, the humanized antibody of the invention or fragment thereof is capable of inhibiting the aggregation of β-amyloid monomers to high molecular polymeric fibrils and of disaggregating preformed polymeric fibrils or filaments.

[0038] In some embodiments, the humanized antibody of the invention or fragment thereof binds to an Aβ fiber, fibril or filament with a binding affinity that is at least 10 times, at least 15 times, at least 20 times, or at least 25 times, higher than the binding affinity of said antibody or fragment thereof to an Aβ monomer.

[0039] In some embodiments, the humanized antibody of the invention or fragment thereof has:
  1. (a) the amino acid in Kabat position 47 of the framework regions of the Heavy Chain Variable Region is the amino acid Leu; and/or
  2. (b) the amino acid in Kabat position 94 in the framework regions of the Heavy Chain Variable Region is the amino acid Ser; and/or
  3. (c) the amino acid in Kabat position 87 in the framework regions of the Light Chain Variable Region is the amino acid Tyr, Phe, Leu, VaI, Ile, or Ala.


[0040] The invention further provides a nucleic acid molecule comprising a nucleotide sequence encoding the humanized antibody of the invention or fragment thereof.

[0041] The invention further provides an expression vector comprising the nucleic acid molecule of the invention.

[0042] The invention further provides a cell comprising the expression vector of the invention.

[0043] The invention further provides a composition comprising the humanized antibody of the invention or fragment thereof, and further comprising a pharmaceutically acceptable carrier, a diluent, and/or an excipient.

[0044] The invention further provides a mixture comprising the humanized antibody of the invention or fragment thereof, and further comprising a biologically active substance, a pharmaceutically acceptable carrier, a diluent, and/or an excipient.

[0045] In some embodiments, the mixture comprises:
  1. (i) a biologically active substance that is a compound used in the treatment of amyloidosis, or
  2. (ii) at least one of the following compounds: an anti-oxidative stress compound; an anti-apoptotic compound; a metal chelator; a DNA repair inhibitor such as pirenzepine and metabolites; 3-amino-1-propanesulfonic acid (3 APS); 1,3-propanedisulfonate (1,3PDS); α-secretase activator; Aβ-secretase inhibitor; a γ-secretase inhibitor; a tau protein; a neurotransmitter; aβ-sheet breaker; an attractant for amyloid beta clearing / depleting cellular components; an inhibitor of N-terminal truncated amyloid beta, such as pyroglutamated amyloid beta 3-42; an anti-inflammatory molecule; a cholinesterase inhibitor (ChEI) such as tacrine, rivastigmine, donepezil, and/or galantamine; an Ml agonist; or another drug such as an amyloid- or tau-modifying drug or nutritive supplement.


[0046] The invention further provides a humanized antibody or fragment thereof as defined in any one of claims 1-9, a composition as defined in claim 13, or a mixture as defined in claim 14 or 15 for use in a method of treating or alleviating the effects of amyloidosis in an animal, such as a mammal or a human.

[0047] In some embodiments, the amyloidosis is secondary amyloidosis, age-related amyloidosis, a neurological disorder, Alzheimer's Disease (AD), Lewy body dementia, Down's syndrome, hereditary cerebral hemorrhage with amyloidosis (Dutch type), the Guam Parkinson-Dementia complex, progressive supranuclear palsy, multiple sclerosis, Creutzfeld Jacob disease, Parkinson's disease, HIV-related dementia, ALS (amyotropic lateral sclerosis), Adult Onset Diabetes, senile cardiac amyloidosis, endocrine tumors, or macular degeneration.

[0048] In some embodiments, the amyloidosis is Alzheimer's disease.

[0049] In some embodiments, the treatment of the animal according to the invention leads to
  1. i) an increase of the cognitive memory capacity and/or;
  2. ii) the retention of cognitive memory capacity; and/or
  3. iii) a complete restoration of the cognitive memory capacity.


[0050] The invention further provides a method of diagnosis of an amyloidosis in a patient comprising:
  1. i) contacting a sample, body part, or body area containing an amyloid protein with the humanized antibody of the invention or fragment thereof under conditions that allow binding of said antibody or fragment thereof to the amyloid protein; and
  2. ii) detecting the antibody or fragment thereof bound to the protein,
wherein the presence or absence of antibody or fragment thereof bound to the amyloid protein indicates the presence or absence of amyloid protein in said sample, body part, or body area.

[0051] The invention further provides a method of determining the extent of amyloidogenic plaque burden in a tissue sample or body fluid sample comprising:
  1. i) testing a tissue sample or body fluid sample for the presence of amyloid protein with the humanized antibody of the invention or fragment thereof;
  2. ii) determining the amount of antibody or fragment thereof bound to the amyloid protein; and
  3. iii) calculating the plaque burden in the tissue sample or body fluid sample.


[0052] The invention further provides a kit for detection and diagnosis of amyloid-associated diseases and conditions comprising the humanized antibody of the invention or fragment thereof, wherein said kit comprises a container holding one or more antibodies of the invention or fragments thereof and instructions for using said antibodies or fragments thereof.

[0053] The invention further provides a humanized antibody of the invention or fragment thereof, a composition of the invention, or a mixture of the invention, for use in a method of preventing degeneration of neurons upon exposure to β-amyloid oligomers.

[0054] The invention further provides a humanized antibody of the invention or fragment thereof, a composition of the invention, or a mixture of the invention, for use in a method of protecting neurons from β-amyloid-induced degradation.

[0055] The invention further provides a method for preparing a humanized antibody of the invention or fragment thereof, wherein the method comprises culturing the cell of the invention, and purifying the antibody or fragment thereof.

SUMMARY OF THE INVENTION



[0056] In one embodiment, the invention relates to a humanized antibody or a fragment thereof, which recognizes and binds to at least one distinct binding site, particularly to a least two distinct binding sites, and more particularly to at least three distinct binding sites on the β-amyloid protein wherein said one, said at least two and said at least three binding sites each comprise at least one or two consecutive amino acid residues predominantly involved in the binding of the antibody.

[0057] In particular, the humanized antibody or a fragment thereof described therein binds to at least two, particularly to at least three distinct binding sites on the β-amyloid protein wherein at least two of the three distinct binding sites comprise at least two consecutive amino acid residues predominantly involved in the binding of the antibody and at least one of the three distinct binding sites comprise at least one amino acid residue.

[0058] The at least two distinct binding sites comprising at least two consecutive amino acid residues predominantly involved in the binding of the antibody are located in close proximity to each other on the antigen, separated and/or flanked by at least one amino acid residue not involved in antibody binding or to a significantly smaller extent as compared to said at least two consecutive amino acid residues, thus forming a conformational discontinuous epitope.

[0059] The at least three distinct binding sites comprising at least two consecutive amino acid residues and at least one amino acid residue, respectively, which are predominantly involved in the binding of the antibody are located in close proximity to each other on the epitope, separated and/or flanked by at least one amino acid residue not involved in antibody binding or to a significantly smaller extent as compared to the amino acid residues, which are predominantly involved in the binding of the antibody, thus forming a conformational discontinuous epitope.

[0060] In particular, a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof is disclosed, which recognizes and binds to at least one distinct binding site, particularly to a least two distinct binding sites, more particularly to at least three distinct binding sites on the β-amyloid- protein wherein said at least one or said at least two distinct binding sites each comprise at least two consecutive amino acid residues predominantly involved in the binding of the antibody, wherein the at least two consecutive amino acid residues representing a first binding site are -Phe-Phe- embedded within the following core sequence (SEQ ID NO: 9):

Xaa3 - Phe - Phe - Xaa4 - Xaa5 - Xaa6, wherein

Xaa3 is an amino acid residue selected from the group consisting of Ala, Val, Leu, norleucine, Met, Phe, and Ile;

Xaa4 is an amino acid residue selected from the group consisting of Ala, Val, Leu, Ser and Ile;

Xaa5 is an amino acid residue selected from the group consisting of Glu and Asp,

Xaa6 is an amino acid residue selected from the group consisting of Glu and Asp, and wherein said amino acid residues Xaa3 Xaa4, Xaa5 and Xaa6 are not involved in antibody binding or to a significantly smaller extent as compared to the -Phe-Phe- binding site.



[0061] Also disclosed is a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof, wherein

Xaa3 is Val or Leu, but particularly Val;

Xaa4 is Ala or Val, but particularly Ala;

Xaa5 is Glu or Asp, but particularly Glu;

Xaa6 is Glu or Asp, but particularly Asp.



[0062] In particular, a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof is disclosed, which recognizes and binds to at least one distinct binding site, particularly to a least two distinct binding sites, more particularly to at least three distinct binding sites on the β-amyloid protein wherein said distinct binding sites comprise at least one and at least two consecutive amino acid residues, respectively, predominantly involved in the binding of the antibody, wherein the at least two consecutive amino acid residues representing a first binding site are -Phe-Phe- and the at least one amino acid residue is -His- embedded within the following core sequence:

- Xaa1 - His - Xaa3 - Xaa4 - Xaa5 - Xaa6 - Phe - Phe - Xaa7 -Xaa8- Xaa9-, wherein

Xaa1 is an amino acid residue selected from the group consisting of His, Asn, Gln, Lys and Arg

Xaa3 is an amino acid residue selected from the group consisting of Asn and Gln

Xaa4 is an amino acid residue selected from the group consisting of His, Asn, Gln, Lys and Arg

Xaa5 is an amino acid residue selected from the group consisting of Ala, Val, Leu, Ser and Ile;

Xaa6 is an amino acid residue selected from the group consisting of Ala, Val, Leu, norleucine, Met, Phe, and Ile

Xaa7 is an amino acid residue selected from the group consisting of Ala, Val, Leu and Ile Xaa8 is an amino acid residue selected from the group consisting of Glu and Asp,

Xaa9 is an amino acid residue selected from the group consisting of Glu and Asp, and wherein said amino acid residues Xaa1, Xaa3, Xaa6, Xaa7, Xaa8 and Xaa9, are not involved in antibody binding or to a smaller to significantly smaller extent as compared to the -His- and the -Phe-Phe- binding site, respectively..



[0063] Also disclosed is a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof, wherein

Xaa3 is Gln or Asn, but particularly Gln;

Xaa4 is Lys

Xaa5 is Leu

Xaa6 is Val or Leu, but particularly Val;

Xaa7 is Ala or Val, but particularly Ala;

Xaa8 is Glu or Asp, but particularly Glu; and

Xaa9 is Asp or Glu, but particularly Asp.



[0064] Also disclosed is a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof, which recognizes and binds to at least one distinct binding site, particularly to a least two distinct binding sites, more particularly to at least three distinct binding sites on the β-amyloid protein, wherein said at least one or said at least two distinct binding sites each comprise at least two consecutive amino acid residues predominantly involved in the binding of the antibody, wherein the at least two consecutive amino acid residues representing a second binding site are -Lys-Leu- embedded within the following core sequence (SEQ ID NO: 10):

Xaa1 - Xaa2 - Lys - Leu - Xaa3 wherein

Xaa1 is an amino acid residue selected from the group consisting of His, Asn, Gln Lys, and Arg;

Xaa2 is an amino acid residue selected from the group consisting of Asn and Gln;

Xaa3 is an amino acid residue selected from the group consisting of Ala, Val, Leu, norleucine, Met, Phe, and Ile; and wherein said amino acid residues Xaa2, Xaa3, are not involved in antibody binding or to a smaller to significantly smaller extent as compared to the -Lys-Leu- binding site.



[0065] Also disclosed is a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof, which recognizes and binds to at least one distinct binding site, particularly to a least two distinct binding sites, more particularly to at least three distinct binding sites on the β-amyloid protein wherein said distinct binding sites comprise at least one and at least two consecutive amino acid residues, respectively, predominantly involved in the binding of the antibody, wherein the at least one and the at least two consecutive amino acids, which are separated by at least one amino acid residue not involved in antibody binding or to a significantly smaller extent as compared to the amino acid residues predominantly involved in the binding of the antibody, are -His- and -Lys-Leu-, respectively, embedded within the following core sequence:

His - Xaa2 - Lys - Leu - Xaa3- Xaa4- Xaa5-Xaa6- - Xaa7 - Xaa8 - wherein

Xaa2 is an amino acid residue selected from the group consisting of Asn and Gln;

Xaa3 is an amino acid residue selected from the group consisting of Ala, Val, Leu, norleucine, Met, Phe, and Ile;

Xaa4 is an amino acid residue selected from the group consisting of Ala, Val, Leu, norleucine, Met, Phe, and Ile

Xaa5 is an amino acid residue selected from the group consisting of Ala, Val, Leu, norleucine, Met, Phe, and Ile

Xaa6 is an amino acid residue selected from the group consisting of Ala, Val, Leu, Ser and Ile;

Xaa7 is an amino acid residue selected from the group consisting of Glu and Asp,

Xaa8 is an amino acid residue selected from the group consisting of Glu and Asp

and wherein said amino acid residues Xaa2, Xaa3, Xaa6, Xaa7, Xaa8, are not involved in antibody binding or to a smaller to significantly smaller extent as compared to the -His- and the -Lys-Leu- binding site, respectively.



[0066] Also disclosed is a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof, wherein

Xaa2 is Gln or Asn, but particularly Gln;

Xaa3 is Val or Leu, but particularly Val;

Xaa4 is Phe

Xaa5 is Phe

Xaa6 is Ala or Val, but particularly Ala;

Xaa7 is Glu or Asp, but particularly Glu; and

Xaa8 is Asp or Glu, but particularly Asp.



[0067] Also disclosed is a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof, which recognizes and binds to at least two distinct binding sites on the β-amyloid protein wherein said at least two distinct binding sites each comprise at least two consecutive amino acid residues predominantly involved in the binding of the antibody, wherein the at least two consecutive amino acids are separated by at least one amino acid residue not involved in antibody binding or to a significantly smaller extent than said consecutive amino acid residues, which are -Phe-Phe- and -Lys-Leu-, respectively, representing a first and second binding site embedded within the following core sequence:

Xaa1 - Xaa2 - Lys - Leu - Xaa3 - Phe - Phe - Xaa4 - Xaa5 - Xaa6, wherein

Xaa1 is an amino acid residue selected from the group consisting of His, Asn, Gln Lys, and Arg;

Xaa2 is an amino acid residue selected from the group consisting of Asn and Gln;

Xaa3 is an amino acid residue selected from the group consisting of Ala, Val, Leu, norleucine, Met, Phe, and Ile;

Xaa4 is an amino acid residue selected from the group consisting of Ala, Val, Leu, Ser and Ile;

Xaa5 is an amino acid residue selected from the group consisting of Glu and Asp,

Xaa6 is an amino acid residue selected from the group consisting of Glu and Asp and wherein said amino acid residues Xaa2, Xaa3, Xaa4, Xaa5 and Xaa6 are not involved in antibody binding or to a smaller to significantly smaller extent as compared to the -Lys-Leu- and - Phe-Phe- binding site, respectively.



[0068] Also disclosed is a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof, which recognizes and binds to at least one distinct binding site, particularly to a least two distinct binding sites, more particularly to at least three distinct binding sites on the β-amyloid protein wherein said distinct binding sites comprise at least one and at least two consecutive amino acid residues, respectively, predominantly involved in the binding of the antibody, wherein the at least one and the at least two consecutive amino acids are separated by at least one amino acid residue not involved in antibody binding or to a significantly smaller extent as compared to the amino acid residues, which are predominantly involved in the binding of the antibody, and wherein said amino acid residues are -His- and -Phe-Phe- and -Lys-Leu-, respectively, embedded within the following core sequence:

His - Xaa2 - Lys - Leu - Xaa3 - Phe - Phe - Xaa4 - Xaa5 - Xaa6, wherein

Xaa2 is an amino acid residue selected from the group consisting of Asn and Gln;

Xaa3 is an amino acid residue selected from the group consisting of Ala, Val, Leu, norleucine, Met, Phe, and Ile;

Xaa4 is an amino acid residue selected from the group consisting of Ala, Val, Leu, Ser and Ile;

Xaa5 is an amino acid residue selected from the group consisting of Glu and Asp,

Xaa6 is an amino acid residue selected from the group consisting of Glu and Asp, and wherein said amino acid residues Xaa2, Xaa3, Xaa4, Xaa5, Xaa6, are not involved in antibody binding or to a smaller to significantly smaller extent as compared to the -His-, the -Lys-Leu- and the -Phe-Phe- binding site, respectively.



[0069] Also disclosed is a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof, wherein

Xaa2 is Gln or Asn, but particularly Gln;

Xaa3 is Val or Leu, but particularly Val;

Xaa4 is Ala or Val, but particularly Ala;

Xaa5 is Glu or Asp, but particularly Glu; and

Xaa6 is Asp or Glu, but particularly Asp.



[0070] Also disclosed is a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof, which recognizes and binds to at least two distinct binding sites on the β-amyloid protein wherein said at least two distinct binding sites each comprise at least two consecutive amino acid residues predominantly involved in the binding of the antibody, wherein the at least two consecutive amino acids are separated by at least one amino acid residue not involved in antibody binding or to a significantly smaller extent than said consecutive amino acid residues, which are -Phe-Phe- and -Lys-Leu-, respectively, representing a first and second binding site embedded within the following core sequence:

Xaa1 - Xaa2 - Lys - Leu - Xaa3 - Phe - Phe - Xaa4 - Xaa5 - Xaa6, wherein

Xaa1 is an amino acid residue selected from the group consisting of His, Asn, Gln, Lys and Arg;

Xaa2 is an amino acid residue selected from the group consisting of Asn and Gln;

Xaa3 is an amino acid residue selected from the group consisting of Val, Ala, Leu, Met, Phe, norleucine and Ile

Xaa4 is an amino acid residue selected from the group consisting of Ala, Val, Leu and Ile;

Xaa5 is an amino acid residue selected from the group consisting of Glu and Asp,

Xaa6 is an amino acid residue selected from the group consisting of Glu and Asp, and wherein said amino acid residues Xaa2, Xaa3, Xaa4, Xaa5, Xaa6, are not involved in antibody binding or to a smaller to significantly smaller extent as compared to the -Lys-Leu- and the -Phe- Phe binding site, respectively.



[0071] Also disclosed is a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof, wherein

Xaa1 is His or Arg, but particularly His;

Xaa2 is Gln or Asn, but particularly Gln;

Xaa3 is Val or Leu, but particularly Val;

Xaa4 is Ala or Val, but particularly Ala;

Xaa5 is Glu or Asp, but particularly Glu; and

Xaa6 is Asp or Glu, but particularly Asp.



[0072] Also disclosed is a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof which recognizes and binds to at least two distinct binding sites on the β-amyloid protein wherein said at least two distinct binding sites each comprise at least two consecutive amino acid residues predominantly involved in the binding of the antibody, which are - Phe - Phe - Ala - Glu -, particularly - Phe - Phe - Ala -,but especially - Phe - Phe - and - Lys - Leu -, respectively, and wherein said at least two distinct binding sites exhibit amino acid sequence -Val - Phe - Phe - Ala - Glu - Asp - shown in SEQ ID NO: 7 and amino acid sequence His - Gln - Lys - Leu - Val - shown in SEQ ID NO: 8, respectively.

[0073] Also disclosed is a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof, which recognizes and binds to at least one distinct binding site, particularly to a least two distinct binding sites, more particularly to at least three distinct binding sites on the β-amyloid protein wherein the said at least one or said at least two distinct binding sites comprise at least one and at least two consecutive amino acid residues, respectively, predominantly involved in the binding of the antibody, which are - Phe - Phe - and - Lys - Leu -, and -His-, respectively, wherein said distinct binding sites are embedded in the amino acid sequence -Val - Phe - Phe - Ala - Glu-, and amino acid sequence -His - Gln - Lys - Leu - Val -, respectively.

[0074] In another aspect the chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof comprises an antigen recognition and binding site which recognizes and binds to at least two distinct binding sites on the β-amyloid protein wherein said at least two distinct binding sites each comprise at least two consecutive amino acid residues within the amino acid sequence given in SEQ ID NOs: 7 and 8, respectively, wherein said consecutive amino acid residues, particularly -Phe- Phe- and -Lys-Leu-, are predominantly involved in the binding of the β-amyloid protein.

[0075] Also disclosed is an antibody or a fragment thereof which binds to 4 distinct binding sites on the β-amyloid protein wherein said 4 distinct binding sites include 2 binding sites each comprising one amino acid residue and 2 binding sites each comprising two consecutive amino acid residues, which residues are predominantly involved in the binding of the antibody, wherein said 4 distinct binding sites are located in close proximity to each other on the β-amyloid protein, and wherein said 4 binding sites are separated by at least one amino acid residue not involved in antibody binding or involved in binding but to a significantly smaller extent as compared to said one amino acid residue and said two consecutive amino acid residues of the 4 distinct binding sites thus forming a conformational discontinuous epitope.

[0076] In particular, the first of the two consecutive amino acid residues predominantly involved in the binding of the antibody is -Lys-Leu-, and the second of the at least two consecutive amino acid residues is -Phe-Phe-, the first of the single amino acid residues is -His- and the second of the single amino acid residues is -Asp- embedded within the following core sequence:
  • Xaa1- His - Xaa2 - Lys - Leu -Xaa3 - Phe - Phe - Xaa4-Xaa5- Asp. - Xaa6 wherein

    Xaa1 is an amino acid residue selected from the group consisting of His, Asn, Gln, Lys and Arg, but particularly His;

    Xaa2 is an amino acid residue selected from the group consisting of Asn and Gln, but particularly Gln ;

    Xaa3 is an amino acid residue selected from the group consisting of Ala, Val, Leu, norleucine, Met, Phe, and Ile, particularly Val;

    Xaa4 is an amino acid residue selected from the group consisting of Ala, Val, Leu, Ser and Ile, particularly Ala;

    Xaa5 is an amino acid residue selected from the group consisting of Glu and Asp, particularly Glu;

    Xaa6 is an amino acid residue selected from the group consisting of Ala, Val, Leu, norleucine, Met, Phe, and Ile, particularly Val; and wherein said amino acid residues Xaa1, Xaa2, Xaa3, Xaa4, Xaa5, Xaa6, are not involved in antibody binding or are involved in binding but to a significantly smaller extent as compared to the -His-, - Asp-, the -Lys-Leu, and the -Phe-Phe- binding site.



[0077] Also disclosed is an antibody or a fragment thereof, which binds to 4 distinct binding sites on the β-amyloid protein, wherein said 4 distinct binding sites include two binding sites each comprising one amino acid residue and two binding sites each comprising two consecutive amino acid residues, wherein the first of the two consecutive amino acid residues predominantly involved in the binding of the antibody is -Lys-Leu-, and the second of the at least two consecutive amino acid residues is -Phe-Phe-, the first of the single amino acid residues is -His- and the second of the single amino acid residues is -Asp- embedded within the following core sequence:

- Xaa1- His - Xaa2 - Lys - Leu -Xaa3 - Phe - Phe - Xaa4- Xaa5- Asp. - Xaa6 wherein

Xaa1 is an amino acid residue selected from the group consisting of His, Asn, Gln, Lys and Arg, but particularly His;

Xaa2 is an amino acid residue selected from the group consisting of Asn and Gln, but particularly Gln ;

Xaa3 is an amino acid residue selected from the group consisting of Ala, Val, Leu, norleucine, Met, Phe, and Ile, particularly Val;

Xaa4 is an amino acid residue selected from the group consisting of Ala, Val, Leu, Ser and Ile, particularly Ala;

Xaa5 is an amino acid residue selected from the group consisting of Glu and Asp, particularly Glu;

Xaa6 is an amino acid residue selected from the group consisting of Ala, Val, Leu, norleucine, Met, Phe, and Ile, particularly Val; and wherein said amino acid residues Xaa1, Xaa2, Xaa3, Xaa4, Xaa5, Xaa6, are not involved in antibody binding or are involved in binding but to a significantly smaller extent as compared to the -His-, - Asp-, the -Lys-Leu, and the -Phe-Phe- binding site.



[0078] In a specific aspect the recognition and binding sites as defined herein before are forming a conformational discontinuous epitope localized in a region of the β-amyloid protein between amino acid residue 12 to 24, particularly between residues 14 to 23, more particularly between amino acid residues 14 and 20, wherein the at least two distinct recognition and binding sites each comprising at least 2 amino acid residues, are located at position 16 and 17 and at position 19 and 20, respectively, and wherein the at least one distinct recognition and binding site comprising at least 1 amino acid residue is located at position 14, which residues are predominantly involved in the binding of the β-amyloid protein and wherein said distinct recognition and binding sites are at least on one side flanked by amino acid residues, particularly residues 21 and 22, and separated by one amino acid residue located at position 15 and 18, which amino acid residues are not directly involved in the binding of the antigen or, at least, to a substantially smaller extent.

[0079] In still another aspect the said at least three distinct recognition and binding sites are flanked on both sides by amino acid residues, particularly residues 12 and 13, and residues 21 and 22 and are separated by one amino acid residue located at position 15 and 18, which amino acid residues are not directly involved in the binding of the antigen or, at least, to a substantially smaller extent.

[0080] In a specific aspect, said consecutive amino acid residues, particularly -Lys-Leu- at position 16 and 17 and -Phe- Phe- at position 19 and 20, which are predominantly involved in the binding of the β-amyloid protein, are embedded into the following core region:
Val- His- His- Gln- Lys- Leu- Val- Phe- Phe- Ala- Glu- Asp
12 13 14 15 16 17 18 19 20 21 22 23


[0081] In another specific aspect, said amino acid residues, particularly -Lys-Leu- at position 16 and 17 and -Phe- Phe- at position 19 and 20, and -His- at position 14, which are predominantly involved in the binding of the β-amyloid protein, are embedded into the following core region:
Val- His- His Gln- Lys- Leu- Val- Phe- Phe- Ala- Glu- Asp- Val- Gly-
12 13 14 15 16 17 18 19 20 21 22 23 24 25


[0082] In another aspect a humanized antibody or a fragment thereof is disclosed which comprises in the light chain and heavy chain variable region, respectively, at least one CDR of non-human origin, particularly two CDRs of non-human origin, more particularly three CDR of non-human origin, embedded in one or more human- or primate-derived framework regions and, optionally, a constant region derived from a human or primate source antibody, which humanized antibody or fragment thereof is capable of specifically recognizing and binding β-amyloid protein, particularly a β-amyloid monomeric peptide, more particularly a β-amyloid polymeric peptide, even more particularly β-amyloid fibers, fibrils or filaments in isolation or as part of a β-amyloid plaque, at an epitope comprising the following amino acid sequence (SEQ ID NO: 11):

Xaa1 - Xaa2 - Lys - Leu - Xaa3 - Phe - Phe- Xaa4 - Xaa5 - Xaa6, wherein

Xaa1 is an amino acid residue selected from the group consisting of His, Asn, Gln, but particularly His;

Xaa2 is an amino acid residue selected from the group consisting of Asn and Gln, but particularly Gln; and

Xaa3 is an amino acid residue selected from the group consisting of Val, Leu, and Ile, but particularly Val;

Xaa4 is an amino acid residue selected from the group consisting of Ala and Val, but particularly Ala;

Xaa5 is an amino acid residue selected from the group consisting of Glu and Asp, but particularly Glu;

Xaa6 is an amino acid residue selected from the group consisting of Glu and Asp, but particularly Asp.



[0083] In still another aspect, a humanized antibody or a fragment thereof is disclosed which comprises in the light chain and heavy chain variable region, respectively, at least one CDR of non-human origin, particularly two CDRs of non-human origin, more particularly three CDR of non-human origin, embedded in one or more human- or primate-derived framework regions and, optionally, a constant region derived from a human or primate source antibody, which humanized antibody or fragment thereof is capable of specifically recognizing and binding β-amyloid protein, particularly a β-amyloid monomeric peptide, more particularly a β-amyloid polymeric peptide, even more particularly β-amyloid fibers, fibrils or filaments in isolation or as part of a β-amyloid plaque, at an epitope comprising the following amino acid sequence:

His - Xaa2 - Lys - Leu - Xaa3 - Phe - Phe- Xaa4 Xaa5 - Xaa6, wherein

Xaa2 is an amino acid residue selected from the group consisting of Asn and Gln, but particularly Gln; and

Xaa3 is an amino acid residue selected from the group consisting of Val, Leu, and Ile, but particularly Val;

Xaa4 is an amino acid residue selected from the group consisting of Ala and Val, but particularly Ala;

Xaa5 is an amino acid residue selected from the group consisting of Glu and Asp, but particularly Glu;

Xaa6 is an amino acid residue selected from the group consisting of Glu and Asp, but particularly Glu; and wherein said amino acid residues Xaa2, Xaa3, Xaa4, Xaa5, Xaa6, are not involved in antibody binding or to a smaller extent as compared to the -His- and the -Lys-Leu- and the -Phe-Phe- binding site.



[0084] In a specific aspect the CDR of non-human origin is obtained from a donor antibody, but particularly from a murine donor antibody, raised against an antigen fragment which does not contain said distinct binding site. This shift in the epitopic region may have at least partially been caused by the use of a supramolecular antigenic construct comprising an antigenic peptide corresponding to the amino acid sequence of the β-amyloid peptide, particularly of β-amyloid peptide Aβ1-16, modified with a hydrophilic moiety such as, for example, polyethylene glycol (PEG), wherein said hydrophilic moiety is covalently bound to each of the termini of the antigenic peptide through at least one, particularly one or two amino acids such as, for example, lysine, glutamic acid and cysteine or any other suitable amino acid or amino acid analogue capable of serving as a connecting device for coupling the hydrophilic moiety to the peptide fragment, as described herein below in the immunization process. When a PEG is used as the hydrophilic moiety, the free PEG termini are covalently bound to phosphatidylethanolamine or any other compound suitable to function as the anchoring element, for example, to embed the antigenic construct in the bilayer of a liposome as described herein.

[0085] In particular, the CDR of non-human origin is obtained from a murine donor antibody which exhibits the characteristic properties of ACI-01-Ab7C2 (also named "mC2" throughout the application) deposited 01 December 2005 with the "Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) in Braunschweig, Mascheroder Weg 1 B, 38124 Branuschweig, under the provisions of the Budapest Treaty under accession no DSM ACC2750).

[0086] In one embodiment of the invention, the CDR of non-human origin is obtained from murine donor antibody ACI-01-Ab7C2 (also named "mC2" throughout the application) deposited 01 December 2005 with the "Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) in Braunschweig, Mascheroder Weg 1 B, 38124 Branuschweig, under the provisions of the Budapest Treaty under accession no DSM ACC2750).

[0087] Also the use of lipid A as part of the immunization protocol may have contributed to a shift in the epitopic region.

[0088] Also disclosed is a humanized antibody or a fragment thereof comprising integrated into human- or primate-derived framework regions at least one peptide with an amino acid sequence selected from the group of sequences consisting of SEQ ID NO: 2 representing CDR2 and SEQ ID NO: 3 representing CDR3 of the Heavy Chain Variable Region (HCVR) and SEQ ID NO: 4 representing CDR1 of the Light Chain Variable Region (LCVR).

[0089] Also disclosed is a humanized antibody or a fragment thereof, wherein said humanized antibody comprises integrated into human- or primate-derived heavy chain framework regions at least one peptide with an amino acid sequence selected from the group of sequences consisting of SEQ ID NO: 2 representing CDR2 and SEQ ID NO: 3 representing CDR3 of the Heavy Chain Variable Region (HCVR). Also disclosed is a humanized antibody or a fragment thereof, wherein said humanized antibody comprises integrated into human- or primate-derived light chain framework regions a peptide with an amino acid sequence of SEQ ID NO: 4 representing CDR1 of the Light Chain Variable Region (LCVR).

[0090] Also disclosed is a Light Chain Variable Region (LCVR) comprising integrated into human- or primate-derived framework regions at least one peptide with an amino acid sequence of SEQ ID NO: 4 representing CDR1 of the Light Chain Variable Region (LCVR).

[0091] Also disclosed is a Heavy Chain Variable Region (HCVR) comprising integrated into human- or primate-derived framework regions at least one peptide with an amino acid sequence selected from the group of sequences consisting of SEQ ID NO: 2 representing CDR2 and SEQ ID NO: 3 representing CDR3 of the Heavy Chain Variable Region (HCVR).

[0092] Also disclosed is a humanized antibody or a fragment thereof, which comprises integrated into human- or primate-derived framework regions at least two peptides, which peptides are different and exhibit an amino acid sequence selected from the group of sequences consisting of SEQ ID NO:1 representing CDR1, SEQ ID NO: 2 representing CDR2 and SEQ ID NO: 3 representing CDR3 of the Heavy Chain Variable Region (HCVR) and SEQ ID NO: 4 representing CDR1, SEQ ID NO: 5 representing CDR2 and SEQ ID NO: 6 representing CDR3 of the Light Chain Variable Region (LCVR) wherein the same CDR cannot be present twice in the antibody. In particular, if the at least two CDRs present are both CDRs of the Light Chain Variable Region (LCVR), at least on of said CDRs must be CDR1 represented by SEQ ID NO: 4.

[0093] Also disclosed is a humanized antibody or a fragment thereof comprising integrated into human- or primate-derived heavy chain framework regions at least two peptides with an amino acid sequence selected from the group of sequences consisting of SEQ ID NO: 1 representing CDR1, SEQ ID NO: 2 representing CDR2 and SEQ ID NO: 3 representing CDR3 of the Heavy Chain Variable Region (HCVR), but particularly a humanized antibody or a fragment thereof wherein the same CDR cannot be present twice in the antibody.

[0094] Also disclosed is a Heavy Chain Variable Region (HCVR) comprising integrated into human- or primate-derived heavy chain framework regions at least two peptides with an amino acid sequence selected from the group of sequences consisting of SEQ ID NO: 1 representing CDR1, SEQ ID NO: 2 representing CDR2 and SEQ ID NO: 3 representing CDR3 of the Heavy Chain Variable Region (HCVR).

[0095] Also disclosed is a humanized antibody or a fragment thereof, comprising integrated into human- or primate-derived light chain framework regions at least two peptides with an amino acid sequence selected from the group of sequences consisting of SEQ ID NO: 4 representing CDR1, SEQ ID NO: 5 representing CDR2 and SEQ ID NO: 6 representing CDR3 of the Light Chain Variable Region (LCVR).

[0096] Also disclosed is a Light Chain Variable Region (LCVR), which has integrated into human- or primate-derived light chain framework regions at least two peptides with an amino acid sequence selected from the group of sequences consisting of SEQ ID NO: 4 representing CDR1, SEQ ID NO: 5 representing CDR2 and SEQ ID NO: 6 representing CDR3 of the Light Chain Variable Region (LCVR), wherein the same CDR cannot be present twice in the antibody and, in particular, at least on of said CDRs must be CDR1 represented by SEQ ID NO: 4.

[0097] Also disclosed is a humanized antibody or a fragment thereof, comprising integrated into human- or primate-derived heavy chain framework regions peptides with an amino acid sequence of SEQ ID NO: 1 representing CDR1, SEQ ID NO: 2 representing CDR2 and SEQ ID NO: 3 representing CDR3 of the Heavy Chain Variable Region (HCVR), particularly in the order indicated above.

[0098] Also disclosed is a Heavy Chain Variable Region (HCVR) comprising integrated into human- or primate-derived heavy chain framework regions peptides with an amino acid sequence of SEQ ID NO: 1 representing CDR1, SEQ ID NO: 2 representing CDR2 and SEQ ID NO: 3 representing CDR3 of the Heavy Chain Variable Region (HCVR), particularly in the order indicated above.

[0099] Also disclosed is a humanized antibody or a fragment thereof comprising integrated into human- or primate-derived light chain framework regions peptides with an amino acid sequence of SEQ ID NO: 4 representing CDR1, SEQ ID NO: 5 representing CDR2 and SEQ ID NO: 6 representing CDR3 of the Light Chain Variable Region (LCVR), particularly in the order indicated above.

[0100] Also disclosed is a Light Chain Variable Region (LCVR) comprising integrated into human- or primate-derived light chain framework regions peptides with an amino acid sequence of SEQ ID NO: 4 representing CDR1, SEQ ID NO: 5 representing CDR2 and SEQ ID NO: 6 representing CDR3 of the Light Chain Variable Region (LCVR), particularly in the order indicated above.

[0101] Also disclosed is a humanized antibody or a fragment thereof, which comprises integrated into human- or primate-derived framework regions at least three peptides with an amino acid sequence selected from the group of sequences consisting of SEQ ID NO: 1 representing CDR1 , SEQ ID NO: 2 representing CDR2 and SEQ ID NO: 3 representing CDR3 of the Heavy Chain Variable Region (HCVR) and SEQ ID NO: 4 representing CDR1, SEQ ID NO: 5 representing CDR2 and SEQ ID NO: 6 representing CDR3 of the Light Chain Variable Region (LCVR), but particularly a humanized antibody or a fragment thereof wherein the same CDR cannot be present twice in the antibody.

[0102] Also disclosed is a humanized antibody or a fragment thereof, which antibody comprises integrated into human- or primate-derived framework regions at least four peptides with an amino acid sequence selected from the group of sequences consisting of SEQ ID NO: 1 representing CDR1, SEQ ID NO: 2 representing CDR2 and SEQ ID NO:3 representing CDR3 of the Heavy Chain Variable Region (HCVR) and SEQ ID NO: 4 representing CDR1, SEQ ID NO: 5 representing CDR2 and SEQ ID NO: 6 representing CDR3 of the Light Chain Variable Region (LCVR), but particularly a humanized antibody or a fragment thereof wherein the same CDR cannot be present twice in the antibody.

[0103] Also disclosed is a humanized antibody or a fragment thereof, which comprises integrated into human- or primate-derived framework regions at least five peptides with an amino acid sequence selected from the group of sequences consisting of SEQ ID NO: 1 representing CDR1, SEQ ID NO: 2 representing CDR2 and SEQ ID NO:3 representing CDR3 of the Heavy Chain Variable Region (HCVR) and SEQ ID NO: 4 representing CDR1, SEQ ID NO: 5 representing CDR2 and SEQ ID NO: 6 representing CDR3 of the Light Chain Variable Region (LCVR), but particularly a humanized antibody or a fragment thereof wherein the same CDR cannot be present twice in the antibody.

[0104] Also disclosed is a humanized antibody or a fragment thereof, which comprises integrated into human- or primate-derived framework regions peptides with an amino acid sequence of SEQ ID NO: 1 representing CDR1, SEQ ID NO: 2 representing CDR2 and SEQ ID NO: 3 representing CDR3 of the Heavy Chain Variable Region (HCVR) and SEQ ID NO: 4 representing CDR1, SEQ ID NO: 5 representing CDR2 and SEQ ID NO: 6 representing CDR3 of the Light Chain Variable Region (LCVR).

[0105] Also disclosed is a antibody, a Heavy Chain Variable Region (HCVR), or a fragment thereof, wherein said humanized antibody, Heavy Chain Variable Region (HCVR) or fragment thereof comprises integrated into human- or primate-derived heavy chain framework regions at least a peptide with an amino acid sequence of SEQ ID NO: 2 representing CDR2 of the Heavy Chain Variable Region (HCVR).

[0106] Also disclosed is a humanized antibody, a Heavy Chain Variable Region (HCVR) or a fragment thereof, wherein said humanized antibody, Heavy Chain Variable Region (HCVR) or fragment thereof comprises integrated into human- or primate-derived heavy chain framework regions at least a peptide with an amino acid sequence of SEQ ID NO: 3 representing CDR3 of the Heavy Chain Variable Region (HCVR).

[0107] Also disclosed is humanized antibody, Heavy Chain Variable Region (HCVR) or a fragment thereof, which antibody, Heavy Chain Variable Region (HCVR) or fragment thereof comprises integrated into human- or primate-derived heavy chain framework regions at least two peptides with an amino acid sequence of SEQ ID NO: 1 representing CDR1 and SEQ ID NO: 2 representing CDR2 of the Heavy Chain Variable Region (HCVR).

[0108] Also disclosed is a humanized antibody, a Heavy Chain Variable Region (HCVR) or a fragment thereof, which antibody, Heavy Chain Variable Region (HCVR) or fragment thereof comprises integrated into human- or primate-derived heavy chain framework regions at least two peptides with an amino acid sequence of SEQ ID NO: 1 representing CDR1 and SEQ ID NO: 3 representing CDR3 of the Heavy Chain Variable Region (HCVR).

[0109] Also disclosed is a humanized antibody, a Heavy Chain Variable Region (HCVR) or a fragment thereof, which antibody, Heavy Chain Variable Region (HCVR) or fragment thereof comprises integrated into human- or primate-derived heavy chain framework regions at least two peptides with an amino acid sequence of SEQ ID NO: 2 representing CDR2 and SEQ ID NO: 3 representing CDR3 of the Heavy Chain Variable Region (HCVR).

[0110] Also disclosed is a humanized antibody, a Light Chain Variable Region (LCVR) or a fragment thereof, which antibody, Light Chain Variable Region (LCVR) or fragment thereof comprises integrated into human- or primate-derived heavy chain framework regions at least two peptides with an amino acid sequence of SEQ ID NO: 4 representing CDR1 and SEQ ID NO: 5 representing CDR2 of the Light Chain Variable Region (LCVR).

[0111] Also disclosed is a humanized antibody, a Light Chain Variable Region (LCVR) or a fragment thereof, which antibody, Light Chain Variable Region (LCVR) or fragment thereof comprises integrated into human- or primate-derived heavy chain framework regions at least two peptides with an amino acid sequence of SEQ ID NO: 4 representing CDR1 and SEQ ID NO: 6 representing CDR3 of the Light Chain Variable Region (LCVR).

[0112] Also disclosed is a humanized antibody or a fragment thereof, wherein both the Heavy Chain Variable Region (HCVR) and the Light Chain Variable Region (LCVR) of the mouse C2 antibody each contributes at least one of its CDR regions to the at least two CDR regions of the humanized antibody. The resulting humanized antibody or a fragment thereof thus may comprise
  • at least an amino acid sequence of SEQ ID NO: 1 representing CDR1 (HCVR) in combination with an amino acid sequence of SEQ ID NO: 4 representing CDR1 (LCVR);
  • at least an amino acid sequence of SEQ ID NO: 2 representing CDR2 (HCVR) in combination with an amino acid sequence of SEQ ID NO: 4 representing CDR1 (LCVR);
  • at least an amino acid sequence of SEQ ID NO: 3 representing CDR3 (HCVR) in combination with an amino acid sequence of SEQ ID NO: 4 representing CDR1 (LCVR);
  • at least an amino acid sequence of SEQ ID NO: 1 representing CDR1 (HCVR) in combination with an amino acid sequence of SEQ ID NO: 5 representing CDR2 (LCVR);
  • at least an amino acid sequence of SEQ ID NO: 2 representing CDR2 (HCVR) in combination with an amino acid sequence of SEQ ID NO: 5 representing CDR2 (LCVR);
  • at least an amino acid sequence of SEQ ID NO:2 representing CDR2 (HCVR) in combination with an amino acid sequence of SEQ ID NO: 6 representing CDR3 (LCVR);
  • at least an amino acid sequence of SEQ ID NO:1 representing CDR1 (HCVR) in combination with an amino acid sequence of SEQ ID NO: 6 representing CDR3 (LCVR);
  • at least an amino acid sequence of SEQ ID NO: 3 representing CDR3 (HCVR) in combination with an amino acid sequence of SEQ ID NO: 5 representing CDR2 (LCVR);
  • at least an amino acid sequence of SEQ ID NO: 3 representing CDR3 (HCVR) in combination with an amino acid sequence of SEQ ID NO: 6 representing CDR3 (LCVR).


[0113] The humanized antibody of the invention may comprise a light chain and/or a heavy chain constant region of human or primate origin.

[0114] In a further embodiment, the invention relates to a humanized antibody or a fragment thereof, wherein at least one, particularly at least one but not more than 5, more particularly at least one but not more than 4, even more particularly at least one but not more than 3, but especially at least one but not more than 2, of the amino acids representative of the light chain and/or heavy chain CDR regions as given in SEQ ID NOs: 1 - 6 is changed through a conservative substitution such that the antibody maintains its full functionality.

[0115] In particular, the invention relates to a humanized antibody or a fragment thereof, wherein in CDR2 of the light chain variable region (LCVR) as given in SEQ ID NO: 5, the Lys at Kabat position 50 is replaced by an amino acid residue selected from the group consisting of Arg, Gln and Glu, particularly by Arg.

[0116] In particular, the invention relates to a light chain variable region (LCVR) wherein in CDR2 as given in SEQ ID NO: 5, the Lys at Kabat position 50 is replaced by an amino acid residue selected from the group consisting of Arg, Gln and Glu, particularly by Arg.
In another embodiment, the invention relates to a humanized antibody or a fragment thereof, wherein in CDR2 of the light chain variable region (LCVR) as given in SEQ ID NO: 5, the Ser at Kabat position 53 is replaced by an amino acid residue selected from the group consisting of Asn or Thr, but particularly by Asn.

[0117] In particular, the invention relates to a light chain variable region (LCVR) wherein in CDR2 as given in SEQ ID NO: 5, the Ser at Kabat position 53 is replaced by an amino acid residue selected from the group consisting of Asn or Thr, but particularly by Asn.

[0118] In one embodiment of the invention, a humanized antibody or a fragment thereof is provided, wherein the Heavy Chain Variable Region (HCVR) has an amino acid sequence that is 90%, particularly 95%, more particularly 98% identical to the sequence given in SEQ ID NO: 15 and 16, respectively.

[0119] In another embodiment of the invention, a humanized antibody or a fragment thereof is provided, wherein the Light Chain Variable Region (LCVR) has an amino acid sequence that is 90%, particularly 95%, more particularly 98% identical to the sequence given in SEQ ID NO: 12 and 13, respectively.
In still another embodiment of the invention, a humanized antibody or a fragment thereof is provided, wherein at least two, but especially three, of the CDR regions of the Heavy Chain Variable Region (HCVR) have an amino acid sequence that is 90%, particularly 95%, more particularly 98% identical to the corresponding CDR region as given in SEQ ID NO: 1 - 3.

[0120] In a further embodiment of the invention, a humanized antibody or a fragment thereof is provided, wherein at least two, but especially three, of the CDR regions of the Light Chain Variable Region (LCVR) have an amino acid sequence that is 90%, particularly 95%, more particularly 98% identical to the corresponding CDR region as given in SEQ ID NO: 4 - 6.

[0121] In still another embodiment, the invention relates to a humanized antibody or a fragment thereof according to the present invention as described herein before wherein the Heavy Chain Variable Region (HCVR) has an amino acid sequence that is 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the sequence given in SEQ ID NO: 15 and 16, respectively.

[0122] In still another embodiment, the invention relates to a humanized antibody or a fragment thereof according to the present invention as described herein before wherein the Light Chain Variable Region (LCVR) has an amino acid sequence that is 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the sequence given in SEQ ID NO: 12 and 13, respectively.

[0123] In still another embodiment, the invention relates to a humanized antibody or a fragment thereof according to the present invention as described herein before, wherein at least one, particularly at least two, but especially three, of the CDR regions of the Heavy Chain Variable Region (HCVR) have an amino acid sequence that is 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the corresponding CDR region as given in SEQ ID NO: 1 - 3.

[0124] In still another embodiment, the invention relates to a humanized antibody or a fragment thereof according to the present invention as described herein before, wherein at least one, particularly at least two, but especially three, of the CDR regions of the Light Chain Variable Region (LCVR) have an amino acid sequence that is 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the corresponding CDR region as given in SEQ ID NO: 4 - 6.

[0125] In still another embodiment, the invention relates to a humanized antibody according to the present invention and as described herein before, wherein at least one of the amino acids representative of the acceptor framework sequences obtained from human germline VH and VK sequences, respectively is changed through a substitution to an amino acid from the corresponding region of murine antibody ACI-01-Ab7C2 or a substitution conservative thereto.

[0126] In particular, the invention relates to a Heavy Chain Variable Region and to a humanized antibody comprising this Heavy Chain Variable Region, respectively, wherein the Trp in Kabat position 47 in the acceptor framework sequence obtained from human germline VH sequences of KABAT subgroup VHIII of the Heavy Chain Variable Region is replaced by an amino acid selected from the group consisting of Leu, norleucine, Ile, Val, Met, Ala, and Phe, particularly Leu and Ile, but especially Leu such as shown in SEQ ID NO: 15.

[0127] The invention further relates to a Heavy Chain Variable Region and to a humanized antibody comprising this Heavy Chain Variable Region, respectively, wherein the Arg in Kabat position 94 in the acceptor framework sequence obtained from human germline VH sequences of KABAT subgroup VHIII of the Heavy Chain Variable Region is replaced by an amino acid selected from the group consisting of Ser and Thr, but especially by Ser such as shown in SEQ ID NO: 15.

[0128] In still another embodiment, the invention relates to a Heavy Chain Variable Region and to a humanized antibody comprising this Heavy Chain Variable Region, respectively, wherein the Trp in Kabat position 47 in the acceptor framework sequence obtained from human germline VH sequences of KABAT subgroup VHIII of the Heavy Chain Variable Region is replaced by an amino acid selected from the group consisting of Leu, norleucine, Ile, Val, Met, Ala, and Phe, particularly Leu and Ile, but especially Leu and the Arg in Kabat position 94 is replaced by an amino acid selected from the group consisting of Ser and Thr, but especially by Ser such as shown in SEQ ID NO: 15.

[0129] The invention further relates to a Light Chain Variable Region and to a humanized antibody comprising this Light Chain Variable Region, respectively, wherein the Gln in Kabat position 45 in the acceptor framework sequence obtained from human germline VK sequences of KABAT subgroup VKII of the Light Chain Variable Region is replaced by an amino acid selected from the group consisting of Lys, Arg, Gln, and Asn, particularly by Lys and Arg, but especially by Lys.

[0130] The invention further relates to a Light Chain Variable Region and to a humanized antibody comprising this Light Chain Variable Region, respectively, wherein the Tyr in Kabat position 87 in the acceptor framework sequence obtained from human germline VK sequences of KABAT subgroup VKII of the Light Chain Variable Region is replaced by an amino acid selected from the group consisting of Phe, Leu, Val, Ile, and Ala, particularly by Leu and Phe, but especially by Phe.

[0131] The invention further relates to a Light Chain Variable Region and to a humanized antibody comprising this Light Chain Variable Region, respectively, wherein the Lys in Kabat position 50 in the CDR2 region obtained from a mouse monoclonal antibody, particularly murine antibody ACI-01-Ab7C2, such as shown in SEQ ID NO: 12 is replaced by an Arg.

[0132] In still another embodiment, the invention relates to a Light Chain Variable Region and to a humanized antibody comprising this Light Chain Variable Region, respectively, wherein the Asn in Kabat position 53 in the CDR2 region obtained from a mouse monoclonal antibody, particularly murine antibody ACI-01-Ab7C2, such as shown in SEQ ID NO: 12 is replaced by Ser.

[0133] In still another embodiment, the invention relates to a humanized antibody, wherein the Trp in Kabat position 47 in the acceptor framework sequence obtained from human germline VH sequences of KABAT subgroup VHIII of the Heavy Chain Variable Region is replaced by an amino acid selected from the group consisting of Leu, norleucine, Ile, Val, Met, Ala, and Phe, particularly Leu and Ile, but especially Leu and the Arg in Kabat position 94 in the acceptor framework sequence obtained from human germline VH sequences of KABAT subgroup VHIII of the Heavy Chain Variable Region is replaced by an amino acid selected from the group consisting of Ser and Thr, but especially by Ser as shown in SEQ ID NO: 15, and the Tyr in Kabat position 87 in the acceptor framework sequence obtained from human germline VK sequences of KABAT subgroup VKII of the Light Chain Variable Region is replaced by an amino acid selected from the group consisting of Phe, Leu, Val, Ile, and Ala, particularly by Leu and Phe, but especially by Phe.

[0134] In still another embodiment, the invention relates to a Heavy Chain Variable Region and to a humanized antibody comprising this Heavy Chain Variable Region, respectively, wherein the Trp in Kabat position 47 in the acceptor framework sequence obtained from human germline VH sequences of KABAT subgroup VHIII of the Heavy Chain Variable Region as shown in SEQ ID NO: 15 is replaced by Leu.

[0135] In still another embodiment, the invention relates to a Heavy Chain Variable Region and to a humanized antibody comprising this Heavy Chain Variable Region, respectively, wherein the Arg in Kabat position 94 in the acceptor framework sequence obtained from human germline VH sequences of KABAT subgroup VHIII of the Heavy Chain Variable Region is replaced by Ser such as shown in SEQ ID NO: 15.

[0136] In still another embodiment, the invention relates to a Heavy Chain Variable Region and to a humanized antibody comprising this Heavy Chain Variable Region, respectively,, wherein the Trp in Kabat position 47 in the acceptor framework sequence obtained from human germline VH sequences of KABAT subgroup VHIII of the Heavy Chain Variable Region is replaced by Leu and Ile, but especially Leu and the Arg in Kabat position 94 in the acceptor framework sequence obtained from human germline VH sequences of KABAT subgroup VHIII of the Heavy Chain Variable Region is replaced by Ser such as shown in SEQ ID NO: 15.

[0137] In still another embodiment, the invention relates to a Light Chain Variable Region and to a humanized antibody comprising this Heavy Chain Variable Region, respectively, wherein the Tyr in Kabat position 87 in the acceptor framework sequence obtained from human germline VK sequences of KABAT subgroup VKII of the Light Chain Variable Region is replaced by Phe.

[0138] In still another embodiment, the invention relates to a Heavy Chain Variable Region and to a humanized antibody comprising this Heavy Chain Variable Region, respectively,, wherein the Trp in Kabat position 47 in the acceptor framework sequence obtained from human germline VH sequences of KABAT subgroup VHIII of the Heavy Chain Variable Region is replaced by Leu and Ile, but especially Leu and the Arg in Kabat position 94 in the acceptor framework sequence obtained from human germline VH sequences of KABAT subgroup VHIII of the Heavy Chain Variable Region is replaced by Ser such as shown in SEQ ID NO: 15 and the Tyr in Kabat position 87 in the acceptor framework sequence obtained from human germline VK sequences of KABAT subgroup VKII of the Light Chain Variable Region is replaced by Phe..

[0139] In one embodiment, the invention relates to a Heavy Chain Variable Region and to a humanized antibody comprising this Heavy Chain Variable Region, respectively, wherein the Trp in Kabat position 47 in the acceptor framework sequence obtained from human germline VH sequences of KABAT subgroup VHIII of the Heavy Chain Variable Region is replaced by an amino acid selected from the group consisting of Leu, norleucine, Ile, Val, Met, Ala, and Phe, particularly Leu and Ile, but especially Leu and the Arg in Kabat position 94 is replaced by an amino acid selected from the group consisting of Ser and Thr, but especially by Ser such as shown in SEQ ID NO: 15 and wherein the Lys in Kabat position 50 in the CDR2 region obtained from a mouse monoclonal antibody, particularly murine antibody ACI-01-Ab7C2, is replaced by an amino acid selected from the group consisting of Arg, Gln, His, and Asn, but especially by Arg.

[0140] In one embodiment, the invention relates to a Heavy Chain Variable Region and to a humanized antibody comprising this Heavy Chain Variable Region, respectively, wherein the Trp in Kabat position 47 in the acceptor framework sequence obtained from human germline VH sequences of KABAT subgroup VHIII of the Heavy Chain Variable Region is replaced by an amino acid selected from the group consisting of Leu, norleucine, Ile, Val, Met, Ala, and Phe, particularly Leu and Ile, but especially Leu and the Arg in Kabat position 94 is replaced by an amino acid selected from the group consisting of Ser and Thr, but especially by Ser such as shown in SEQ ID NO: 15 and wherein the Asn in Kabat position 53 in the CDR2 region obtained from a mouse monoclonal antibody, particularly murine antibody ACI-01-Ab7C2, is replaced by an amino acid selected from the group consisting of Ala, Val, Leu, Ser and Ile; but especially Ser.The light chain variable region of SEQ ID NO: 12 is disclosed. In another specific embodiment of the invention, a humanized antibody is provided, which comprises the light chain variable region of SEQ ID NO: 12.

[0141] Disclosed is the light chain variable region including signal sequences as shown in SEQ ID NO: 13.

[0142] In another specific embodiment of the invention, a humanized antibody is provided, which comprises the complete light chain variable region including signal sequences as shown in SEQ ID NO: 13.

[0143] In another specific embodiment of the invention, a humanized antibody is provided, which comprises the light chain variable region of SEQ ID NO: 12 and the light chain constant region of SEQ ID NO: 14.

[0144] In another specific embodiment of the invention, a humanized antibody is provided, which comprises the complete light chain variable region of SEQ ID NO: 13 and the light chain constant region of SEQ ID NO: 14. Disclosed is also the heavy chain variable region of SEQ ID NO: 15.

[0145] In another specific embodiment of the invention, a humanized antibody is provided, which comprises the heavy chain variable region of SEQ ID NO: 15. Disclosed is the heavy chain variable region including signal sequences as shown in SEQ ID NO: 16.

[0146] In another specific embodiment of the invention, a humanized antibody is provided, which comprises the complete heavy chain variable region including signal sequences as shown in SEQ ID NO: 16.
In another specific embodiment of the invention, a humanized antibody is provided, which comprises the heavy chain variable region of SEQ ID NO: 15 and the heavy chain constant region of SEQ ID NO: 17.
In another specific embodiment of the invention, a humanized antibody is provided, which comprises the heavy chain variable region of SEQ ID NO: 16 and the heavy chain constant region of SEQ ID NO: 17.
In one embodiment the humanized antibody according to the invention and as described herein, upon co-incubation with an Aβ monomeric peptide having at least 30, particularly at least 35, more particularly at least 38, even more particularly at least 40 amino acid residues and/or an Aβ polymeric soluble amyloid peptide comprising a plurality of said Aβ monomeric units, but especially with an Aβ1-42 monomeric and/or an Aβ polymeric soluble amyloid peptide comprising a plurality of said Aβ1-42 monomeric units, particularly at a molar concentration ratio of antibody to Aβ1-42 of up to 1:1000, particularly of up to 1:500, more particularly of up to 1:300, even more particularly of up to 1:200, but especially at a molar concentration ratio of between 1:10 and 1:100, inhibits the aggregation of the Aβ monomers to high molecular polymeric fibrils.

[0147] In particular, the co-incubation of the antibody according to the invention with amyloid monomeric and/or polymeric soluble amyloid peptides is carried out for 24 hours to 60 hours, particularly for 30 hours to 50 hours, more particularly for 48 hours, but especially 24 hours, at a temperature of between 28°C and 40°C, particularly of between 32°C and 38°C, more particularly at 37°C.

[0148] In a specific aspect co-incubation with amyloid monomeric and/or polymeric soluble amyloid peptides is accomplished for 24 hours at a temperature of 37°C.

[0149] In particular, the humanized antibody according to the invention including any functional parts thereof binds to Aβ1-42 monomeric peptide and/or Aβ polymeric soluble amyloid peptide comprising a plurality of said Aβ1-42 monomeric units and, upon co-incubation with Aβ1-42 monomeric peptide and/or Aβ polymeric soluble amyloid peptide comprising a plurality of said Aβ1-42 monomeric units inhibits the aggregation of the Aβ monomers and/or polymers to high molecular polymeric fibrils.

[0150] In one embodiment, the humanized antibody according to the invention including any functional parts thereof inhibits the aggregation of the Aβ monomers and/or Aβ soluble polymers comprising a plurality of said Aβ monomeric units to high molecular polymeric fibrils by at least 50%, particularly by at least 60%, particularly by at least 65%, more particularly by at least 75%, even more particularly by at least 80%, but especially by at least 85%-90%, or more as compared to the respective amyloid peptide monomers incubated in buffer (control), at a molar concentration ratio of antibody to Aβ1-42 of up to 1:1000, particularly at a molar concentration ratio of between 1:10 and 1:100, but especially at a molar concentration ratio of 1:10.

[0151] In a specific embodiment of the invention, the humanized antibody according to the invention including any functional parts thereof inhibits the aggregation of the Aβ monomers and/or Aβ soluble polymers comprising a plurality of said Aβ monomeric units to high molecular polymeric fibrils by at least 30% at a molar concentration ratio of antibody to Aβ1-42 of 1:100.

[0152] In another specific embodiment of the invention, the humanized antibody according to the invention including any functional parts thereof inhibits the aggregation of the Aβ monomers and/or Aβ soluble polymers comprising a plurality of said Aβ monomeric units to high molecular polymeric fibrils by at least 80% at a molar concentration ratio of antibody to Aβ1-42 of 1:10.

[0153] Binding of the antibodies according to the invention and as described herein to amyloidogenic monomeric and/or polymeric peptides but, particularly, to the amyloid form (1-42) leads to inhibition of the aggregation of monomeric and/or polymeric amyloidogenic peptides to high molecular fibrils or filaments. Through the inhibition of the aggregation of amyloidogenic monomeric and/or polymeric peptides the antibodies according to the present invention are capable of preventing or slowing down the formation of amyloid plaques, particularly the amyloid form (1-42), which is know to become insoluble by change of secondary conformation and to be the major part of amyloid plaques in brains of diseased animals or humans.

[0154] The aggregation inhibition potential of the antibody according to the invention may be determined by any suitable method known in the art, particularly by density-gradient ultracentrifugation followed by a SDS-PAGE sedimentation analysis on a preformed gradient and/or by a thioflavin T (Th-T) fluorescent assay.

[0155] In one embodiment, the invention relates to a humanized antibody as described herein including any functional parts thereof, which antibody, upon co-incubation, particularly at a molar concentration ratio of between 1:5 and 1:1000, particularly of between 1:10 and 1:500, more particularly at a ratio of 1:10 to 1:300, even more particularly at a ratio of between 1:10 and 1:100, with preformed high molecular polymeric amyloid fibrils or filaments formed by the aggregation of Aβ monomeric peptides having at least 30, particularly at least 35, more particularly at least 38, even more particularly at least 40 amino acid residues and, but especially Aβ1-42 monomeric peptides, is capable of disaggregating the preformed polymeric fibrils or filaments by at least 20%, particularly by at least 30%, more particularly by at least 35%%, even more particularly by at least 40%, but especially by at least 50% or more.

[0156] In a specific embodiment of the invention, the aggregation inhibition and the disaggregation potential of the antibody, respectively, is determined by density-gradient ultracentrifugation followed by a SDS-PAGE sedimentation analysis on a preformed gradient.

[0157] In another specific embodiment of the invention, the aggregation inhibition and the disaggregation potential of the antibody, respectively, is determined by thioflavin T (Th-T) fluorescent assay.

[0158] In another specific embodiment, the antibody according to the invention is co-incubated with amyloid preformed high molecular polymeric amyloid fibrils or filaments for 12 hours to 36 hours, particularly for 18 hours to 30 hours, more particularly for 24 hours at a temperature of between 28°C and 40°C, particularly of between 32°C and 38°C, more particularly at 37°C.

[0159] In particular, the co-incubation with preformed high molecular polymeric amyloid fibrils or filaments is done for 24 hours at a temperature of 37°C.

[0160] In a specific embodiment of the invention, the humanized antibody according to the invention including any functional parts thereof is capable of disaggregating the preformed polymeric fibrils or filaments by at least 24% at a molar concentration ratio of antibody to Aβ1-42 of 1:100.

[0161] In another specific embodiment of the invention, the humanized antibody according to the invention including any functional parts thereof is capable of disaggregating the preformed polymeric fibrils or filaments by at least 32% at a molar concentration ratio of antibody to Aβ1-42 of 1:10.

[0162] Through the disaggregation of amyloidogenic polymeric fibrils or filaments the antibodies according to the present invention are capable of preventing or slowing down the formation of amyloid plaques which leads to an alleviation of the symptoms associated with the disease and a delay or reversal of its progression.

[0163] Accordingly, it is a further embodiment of the invention to provide a humanized antibody, including any functional parts thereof as described herein, which antibody is capable of decreasing the total amount of Aβ in the brain of an animal, particularly a mammal, but especially a human suffering from a disease or condition leading to increased concentration of Aβ in the brain.

[0164] In another embodiment, the invention relates to a humanized antibody according to the invention and as described herein before, which antibody is bi-effective in that it exhibits both an aggregation inhibition property as well as a disaggregation property, particularly paired with a high degree of conformational sensitivity.

[0165] In particular, the invention relates to a humanized antibody or a fragment thereof according to the invention and as described herein before, which antibody, upon co-incubation with amyloid monomeric and/or polymeric soluble amyloid peptides, particularly with β-amyloid monomeric peptides such as, for example, Aβ monomeric peptides 1-39; 1-40, 1-41, or 1-42, and/or a polymeric soluble β-amyloid peptide comprising a plurality of said Aβ monomeric units, but especially with an Aβ1-42 monomeric and/or an Aβ polymeric soluble amyloid peptide comprising a plurality of said Aβ1-42 monomeric units,, inhibits the aggregation of the Aβ monomers into high molecular polymeric fibrils or filaments and, in addition, upon co-incubation with preformed high molecular polymeric amyloid fibrils or filaments formed by the aggregation of amyloid monomeric peptides, particularly β-amyloid monomeric peptides such as, for example, Aβ monomeric peptides 1-39; 1-40, 1-41, or 1-42, but especially Aβ1-42 monomeric peptides, is capable of disaggregating the preformed polymeric fibrils or filaments.

[0166] In another aspect, the invention relates to a humanized antibody or a fragment thereof according to the present invention and as described herein before, which antibody is capable of inducing a transition of the β-sheet conformation towards an α-helix and/or a random coil conformation, but particularly a random coil conformation, even more particularly a random coil conformation at a given location in the molecule, especially in the environment of Tyr 10 and Val12 of the Aβ protein, which leads to an increase of the random coil conformation at the expense of the β-sheet conformation and an improved solubilization of the preformed high molecular polymeric amyloid fibrils or filaments. In particular the decrease of the β-sheet conformation amounts to at least 30%, particularly to at least 35%, and more particularly to at least 40% and more as compared to the respective preformed amyloid polymeric fibrils or filaments incubated in buffer (control).

[0167] The antibody's potential in inducing a transition in the secondary structure is determined by solid state 13C NMR spectroscopy but, in particular, by measuring the integral intensities of the conformations of Tyr 10 and Val 12 Cβ in the Aβ1-42 peptide. A humanized antibody or a fragment as described herein before, is provided comprising at least one light chain or a fragment thereof or at least one heavy chain or a fragment thereof, wherein said antibody or fragment binds to an Aβ monomer with a high binding affinity with a KD in a range of between at least about 1 x 10-7 M to at least about 1 x 10-12 M, particularly of at least about 1 x 10-8 M to at least about 1 x 10-11 M, more particularly of at least about 1 x 10-9 M to at least about 1 x 10-10 M, even more particularly of at least about 1 x 10-8 M to at least about 2 x 10-8.M, but, preferably, does not show any significant cross-reactivity with amyloid precursor protein (APP). A humanized antibody or a fragment as described herein before, is provided comprising at least one light chain or a fragment thereof or at least one heavy chain or a fragment thereof, wherein said antibody or fragment binds to an Aβ fiber, fibril or filament with a high binding affinity with a KD in a range of between at least about 1 x 10-7 M to at least about 1 x 10-12 M, particularly of at least about 1 x 10-8 M to at least about 1 x 10-11 M, more particularly of at least about 1 x 10-9 M to at least about 1 x 10-10 M, even more particularly of at least about 2 x 10-9 M to at least about 5 x 10-9 M, but, preferably, does not show any significant cross-reactivity with amyloid precursor protein (APP).

[0168] In another embodiment, the humanized antibody according to the invention and as described herein before or a fragment thereof, exhibits an binding affinity to an Aβ fiber, fibril or filament which is at least 2 times, particularly at least 4 times, particularly at least 10 times, particularly at least 15 times, more particularly at least 20 times, but especially at least 25 times higher than the binding affinity to an Aβ monomer.

[0169] In still another embodiment, a humanized antibody or a fragment thereof according to the present invention and as described herein before, substantially binds to aggregated Aβ, including Aβ plaques, in the mammalian, particularly the human brain but, preferably, does not show any significant cross-reactivity with amyloid precursor protein (APP). In another aspect of the invention, a humanized antibody or a fragment thereof according to the present invention and as described herein before, substantially binds to soluble polymeric amyloid, particularly amyloid β (Aβ), including Aβ monomers, in the mammalian, particularly the human brain but, preferably, does not show any significant cross-reactivity with amyloid precursor protein (APP).

[0170] Further provided is a humanized antibody or a fragment thereof according to the invention and as described herein before, which antibody significantly reduces Aβ plaque burden in the mammalian, particularly the human brain. This can be achieved by either binding of the antibody to the plaque or by shifting the equilibrium between amyloid, particularly amyloid β (Aβ), in its insoluble and aggregated state towards its soluble form by disaggregating fibers to soluble poly- and monomeric forms by inducing a shift in conformation and binding and stabilizing the disaggregated and solubilized amyloid forms, particularly amyloid β (Aβ) forms, in the tissue and/or body fluids, particularly the brain. Through the activity of the antibody according to the invention the peripheral clearing and catabolism is thus favored rather than deposition within the tissue and/or body fluids, particularly the brain. The beneficial effect of the antibody according to the invention can thus be obtained without binding of the antibody to the plaque.

[0171] Through this stabilizing activity, the antibody according to the invention is able to neutralize the toxic effects of the polymeric and less aggregated soluble amyloid protein, particularly amyloid β (Aβ) protein, in the tissue and/or body fluids. In a specific embodiment of the invention the antibody according to the invention may thus achieve its beneficial effects without necessarily binding aggregated amyloid beta in the brain. A humanized antibody or a fragment thereof as described herein before, is provided comprising at least one light chain or a fragment thereof or at least one heavy chain or a fragment thereof incorporating at least one, particularly two and more particularly three CDR regions obtained form a mouse donor antibody, particularly from mouse antibody ACI-01-Ab7C2 (named "mC2" and hC2 for the humanized C2 antibody, throughout the application) deposited 01 December 2005 with the "Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) in Braunschweig, Mascheroder Weg 1 B, 38124 Braunschweig, under accession no DSM ACC2750, wherein said antibody or fragment thereof has an affinity to the Aβ antigen which is at least 5 times, particularly at least 8 times, more particularly at least 10 times, but especially at least 15 times higher than that of the mouse donor antibody.

[0172] The antibody of this invention can be, in one embodiment, a whole antibody (e.g., with two full length light chains and two full length heavy chains) of any isotype and subtype (e.g., IgM, IgD, IgG1, IgG2, IgG3, IgG4, IgE, IgA1 and IgA2); but especially an antibody of the IgG4 isotype; alternatively, in another embodiment, it can be an antigen-binding fragment (e.g., Fab, F(ab')2, and Fv) of a whole antibody.

[0173] The invention thus also relates to antigen-binding fragments of the antibodies described herein. In one embodiment of the invention, the fragment is selected from the group consisting of a Fab fragment, a Fab' fragment, a F(ab)2fragment, and a Fv fragment, including the products of an Fab immunoglobulin expression library and epitope-binding fragments of any of the antibodies and fragments mentioned above.

[0174] In another embodiment, the antibody or antigen-binding fragment of the invention is conjugated to polyethylene glycol. In yet another embodiment, the constant region of the antibody of the invention is modified to reduce at least one constant region-mediated biological effector function relative to an unmodified antibody. In still another embodiment, the antibody or antigen-binding fragment of the invention comprises a Fc region having an altered effector function.

[0175] The invention further relates to a nucleotide molecule comprising a nucleotide sequence encoding a humanized antibody or a fragment thereof according to the invention and as disclosed herein before.

[0176] In particular, the invention relates to a nucleotide molecule comprising a nucleotide sequence encoding a stretch of contiguous amino acid molecules as given in SEQ ID NO: 2 and 3, respectively, or the complementary sequence, representing the Complementarity Determining Regions (CDRs) 2 and 3 of the Heavy Chain Variable Region (HCVR).

[0177] More particularly, the invention relates to a nucleotide molecule comprising a nucleotide sequence encoding a stretch of contiguous amino acid molecules as given in SEQ ID NO: 4, or the complementary sequence, representing the Complementarity Determining Regions (CDRs) 1 of the Light Chain Variable Region (LCVR).

[0178] In another aspect a nucleotide molecule is provided comprising a nucleotide sequence as given in SEQ ID NO: 18 and SEQ ID NO: 19, or the complementary sequence, encoding the amino acid sequence of CDR 2 and CDR 3, respectively, of the Heavy Chain Variable Region (HCVR).

[0179] In another aspect a nucleotide molecule is provided comprising a nucleotide sequence as given in SEQ ID NO: 20, or the complementary sequence, encoding the nucleotide sequence of CDR 1 of the Light Chain Variable Region (LCVR).

[0180] In another embodiment of the invention the nucleotide molecule of the invention comprises a nucleotide sequence of SEQ ID NO: 21, or the complementary sequence, encoding the light chain variable region.

[0181] In another embodiment of the invention the nucleotide molecule of the invention comprises a nucleotide sequence of SEQ ID NO: 22, or the complementary sequence, encoding the complete light chain variable region including signal sequences.

[0182] In another embodiment of the invention the nucleotide molecule of the invention comprises a nucleotide sequence encoding the light chain variable region of SEQ ID NO: 22 and the light chain constant region of SEQ ID NO: 23.

[0183] In another embodiment of the invention the nucleotide molecule of the invention comprises a nucleotide sequence of SEQ ID NO: 24 encoding the heavy chain variable region.

[0184] In another embodiment of the invention the nucleotide molecule of the invention comprises a nucleotide sequence of SEQ ID NO: 25 encoding the complete heavy chain variable region including signal sequences.

[0185] In another embodiment of the invention a nucleotide molecule is provided comprising a nucleotide sequence encoding the heavy chain variable region of SEQ ID NO: 25 and the heavy chain constant region of SEQ ID NO: 26 . The invention also comprises the complementary strand of said nucleotide molecule.

[0186] Also comprised herein is a nucleotide sequence which hybridizes to one of the above-described antibody-encoding nucleotide sequences of the invention, particularly to the complementary strand thereof, either in isolation or as part of larger nucleotide molecule.

[0187] In particular, the invention relates to a nucleotide sequence that hybridizes under conventional hybridization conditions, particularly under stringent hybridization conditions, to any of the nucleotide sequences given in SEQ ID NOs: 18-26 and 29 - 32, particularly to the complementary strand thereof.

[0188] In another embodiment of the invention an expression vector is provided comprising the nucleic acid molecule according to the invention. In another embodiment of the invention a cell is provided comprising an expression vector comprising the nucleic acid according to the invention. In still another embodiment, the invention relates to a composition comprising the antibody according to the invention, but particularly a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof according to the invention and as described herein before including any functionally equivalent antibody or any derivative or functional parts thereof, in a therapeutically effective amount, in particular a composition which is a pharmaceutical composition optionally further comprising a pharmaceutically acceptable carrier. In another embodiment of the invention, said composition comprises the antibody in a therapeutically effective amount.

[0189] Further comprised by the invention is a mixture comprising a humanized antibody or a fragment thereof according to the invention including any functional parts thereof, in a therapeutically effective amount and, optionally, a further biologically active substance and/or a pharmaceutically acceptable carrier and/or a diluent and/or an excipient.

[0190] In particular, the invention relates to a mixture, wherein the further biologically active substance is a compound used in the medication of amyloidosis, a group of diseases and disorders associated with amyloid or amyloid-like protein such as the Aβ protein involved in Alzheimer's disease.

[0191] In another embodiment of the invention, the other biologically active substance or compound may also be a therapeutic agent that may be used in the treatment of amyloidosis caused by amyloid β or may be used in the medication of other neurological disorders.

[0192] The other biologically active substance or compound may exert its biological effect by the same or a similar mechanism as the antibody according to the invention or by an unrelated mechanism of action or by a multiplicity of related and/or unrelated mechanisms of action.

[0193] Generally, the other biologically active compound may include neutron-transmission enhancers, psychotherapeutic drugs, acetylcholine esterase inhibitors, calcium-channel blockers, biogenic amines, benzodiazepine tranquillizers, acetylcholine synthesis, storage or release enhancers, acetylcholine postsynaptic receptor agonists, monoamine oxidase-A or -B inhibitors, N-methyl-D-aspartate glutamate receptor antagonists, non-steroidal anti-inflammatory drugs, antioxidants, and serotonergic receptor antagonists.

[0194] More particularly, the invention relates to a mixture comprising at least one compound selected from the group consisting of compounds effective against oxidative stress, anti-apoptotic compounds, metal chelators, inhibitors of DNA repair such as pirenzepin and metabolites, 3-amino-1-propanesulfonic acid (3APS), 1,3-propanedisulfonate (1,3PDS), α-secretase activators, β- and γ-secretase inhibitors, tau proteins, neurotransmitter, β-sheet breakers, attractants for amyloid beta clearing / depleting cellular components, inhibitors of N-terminal truncated amyloid beta including pyroglutamated amyloid beta 3-42, anti-inflammatory molecules, or cholinesterase inhibitors (ChEIs) such as tacrine, rivastigmine, donepezil, and/or galantamine, M1 agonists and other drugs including any amyloid or tau modifying drug and nutritive supplements, and nutritive supplements, together with an antibody according to the present invention and, optionally, a pharmaceutically acceptable carrier and/or a diluent and/or an excipient.

[0195] The invention further relates to a mixture, wherein the compound is a cholinesterase inhibitor (ChEIs), particularly a mixture, wherein the compound is one selected from the group consisting of tacrine, rivastigmine, donepezil, galantamine, niacin and memantine.

[0196] In a further embodiment, the mixtures according to the invention may comprise niacin or memantine together with an antibody according to the present invention and, optionally, a pharmaceutically acceptable carrier and/or a diluent and/or an excipient.

[0197] In still another embodiment of the invention mixtures are provided that comprise "atypical antipsychotics" such as, for example clozapine, ziprasidone, risperidone, aripiprazole or olanzapine for the treatment of positive and negative psychotic symptoms including hallucinations, delusions, thought disorders (manifested by marked incoherence, derailment, tangentiality), and bizarre or disorganized behavior, as well as anhedonia, flattened affect, apathy, and social withdrawal, together with an antibody, particularly a monoclonal antibody according to the invention, but particularly a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof according to the invention and as described herein and, optionally, a pharmaceutically acceptable carrier and/or a diluent and/or an excipient.

[0198] In a specific embodiment of the invention, the compositions and mixtures according to the invention comprise the antibody and the biologically active substance, respectively, in a therapeutically effective amount.

[0199] Other compounds that can be suitably used in mixtures in combination with the antibody according to the present invention are described in WO 2004/058258 (see especially pages 16 and 17) including therapeutic drug targets (page 36-39), alkanesulfonic acids and alkanolsulfuric acids (pages 39-51), cholinesterase inhibitors (pages 51-56), NMDA receptor antagonists (pages 56-58), estrogens (pages 58-59), non-steroidal anti-inflammatory drugs (pages 60-61), antioxidants (pages 61-62), peroxisome proliferators-activated receptor (PPAR) agonists (pages 63-67), cholesterol-lowering agents (pages 68-75); amyloid inhibitors (pages 75-77), amyloid formation inhibitors (pages 77-78), metal chelators (pages 78-79), anti-psychotics and antidepressants (pages 80-82), nutritional supplements (pages 83-89) and compounds increasing the availability of biologically active substances in the brain (see pages 89-93) and prodrugs (pages 93 and 94).

[0200] In another embodiment, the invention relates to a mixture comprising the antibody, particularly a monoclonal antibody according to the invention, but particularly a chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof according to the invention and as described herein before and/or the biologically active substance in a therapeutically effective amount.

[0201] The invention further relates to the use of a humanized antibody or a fragment thereof according to the invention and as described herein before and/or a functional part thereof and/or a pharmaceutical composition, or a mixture comprising said antibody, for the preparation of a medicament for treating or alleviating the effects of amyloidosis, a group of diseases and disorders associated with amyloid plaque formation including secondary amyloidosis and age-related amyloidosis such as diseases including, but not limited to, neurological disorders such as Alzheimer's Disease (AD), Lewy body dementia, Down's syndrome, hereditary cerebral hemorrhage with amyloidosis (Dutch type); the Guam Parkinson-Dementia complex; as well as other diseases which are based on or associated with amyloid-like proteins such as progressive supranuclear palsy, multiple sclerosis; Creutzfeld Jacob disease, Parkinson's disease, HIV-related dementia, ALS (amyotropic lateral sclerosis), Adult Onset Diabetes; senile cardiac amyloidosis; endocrine tumors, and others, including macular degeneration.

[0202] Also comprised by the present invention is a method for the preparation of a humanized antibody or a fragment thereof according to the invention and/or a functional part thereof and/or a pharmaceutical composition, or a mixture comprising said antibody and/or a functional part thereof, particularly in a therapeutically effective amount, for use in a method of preventing, treating or alleviating the effects of amyloidosis, a group of diseases and disorders associated with amyloid plaque formation including secondary amyloidosis and age-related amyloidosis such as diseases including, but not limited to, neurological disorders such as Alzheimer's Disease (AD), Lewy body dementia, Down's syndrome, hereditary cerebral hemorrhage with amyloidosis (Dutch type); the Guam Parkinson-Dementia complex; as well as other diseases which are based on or associated with amyloid-like proteins such as progressive supranuclear palsy, multiple sclerosis; Creutzfeld Jacob disease, Parkinson's disease, HIV-related dementia, ALS (amyotropic lateral sclerosis), Adult Onset Diabetes; senile cardiac amyloidosis; endocrine tumors, and others, including macular degeneration comprising formulating a humanized antibody or a fragment thereof according to the invention in a pharmaceutically acceptable form.

[0203] Further comprised by the present invention is a humanized antibody and/or a functional part thereof. or a composition or mixture comprising such an antibody and/or a functional part thereof for use in a method for preventing, treating or alleviating the effects of amyloidosis, a group of diseases and disorders associated with amyloid plaque formation including secondary amyloidosis and age-related amyloidosis such as diseases including, but not limited to, neurological disorders such as Alzheimer's Disease (AD), Lewy body dementia, Down's syndrome, hereditary cerebral hemorrhage with amyloidosis (Dutch type); the Guam Parkinson-Dementia complex; as well as other diseases which are based on or associated with amyloid-like proteins such as progressive supranuclear palsy, multiple sclerosis; Creutzfeld Jacob disease, Parkinson's disease, HIV-related dementia, ALS (amyotropic lateral sclerosis), Adult Onset Diabetes; senile cardiac amyloidosis; endocrine tumors, and others, including macular degeneration comprising administering the antibody in a therapeutically effective amount to an animal or a human affected by such a disorder.

[0204] It is also an object of the invention to treat amyloidosis, a group of diseases and disorders associated with amyloid plaque formation including secondary amyloidosis and age-related amyloidosis including, but not limited to, neurological disorders such as Alzheimer's Disease (AD), particularly a disease or condition characterized by a loss of cognitive memory capacity by administering to an animal, particularly a mammal or a human, a humanized antibody, particularly a pharmaceutical composition according to the invention and as described herein.

[0205] In a specific embodiment the invention relates to retaining or increasing cognitive memory capacity but, particularly, for restoring the cognitive memory capacity of an animal, particularly a mammal or a human, suffering from memory impairment by administering to an animal, particularly a mammal or a human, a humanized antibody, particularly a pharmaceutical composition according to the invention. It is a further object of the invention to provide a therapeutic composition and treatment of amyloidosis, a group of diseases and disorders associated with amyloid plaque formation including secondary amyloidosis and age-related amyloidosis including, but not limited to, neurological disorders such as Alzheimer's Disease (AD), particularly a disease or condition characterized by a loss of cognitive memory capacity, using an antibody according to the invention and as described herein before.

[0206] In particular, the invention relates to the use of a humanized antibody of the invention in a method for the treatment of an animal, particularly a mammal or a human, suffering from an amyloid-associated condition characterized by a loss of cognitive memory capacity leads to the retention of cognitive memory capacity.

[0207] The invention further relates to a method of diagnosis of an amyloid-associated disease or condition in a patient comprising detecting the immunospecific binding of an antibody or an active fragment thereof to an epitope of the amyloid protein in a sample or in situ which includes the steps of
  1. (a) bringing the sample or a specific body part or body area suspected to contain the amyloid protein into contact with a humanized antibody or a fragment thereof according to the invention and/or a functional part thereof, which antibody binds an epitope of the amyloid protein;
  2. (b) allowing the antibody and/or a functional part thereof, to bind to the amyloid protein to form an immunological complex;
  3. (c) detecting the formation of the immunological complex; and
  4. (d) correlating the presence or absence of the immunological complex with the presence or absence of amyloid protein in the sample or specific body part or area.


[0208] The invention also relates to a method of determining the extent of amyloidogenic plaque burden in a tissue and/or body fluids comprising
  1. (a) testing a tissue and/or body fluid sample for the presence of amyloid protein with a humanized antibody or a fragment thereof according to the invention and/or a functional part thereof;
  2. (b) determining the amount of antibody bound to the protein; and
  3. (c) calculating the plaque burden in the tissue or body fluid sample


[0209] In particular, the formation of the immunological complex in step b is determined such that presence or absence of the immunological complex correlates with presence or absence of amyloid protein.

[0210] In another embodiment of the invention, a test kit for detection and diagnosis of amyloid-associated diseases and conditions is provided comprising a humanized antibody or a fragment thereof according to the invention and/or a functional part thereof.

[0211] In particular, the invention relates to a test kit for detection and diagnosis of amyloid-associated diseases and conditions comprising a container holding one or more antibodies according to the present invention, and/or a functional part thereof, and instructions for using the antibodies for the purpose of binding to amyloid protein to form an immunological complex and detecting the formation of the immunological complex such that presence or absence of the immunological complex correlates with presence or absence of amyloid protein.

[0212] In another aspect, disclosed herein is an antibody comprising a variable region as recited in SEQ ID NO: 27, or a variant thereof. In one aspect, a cell line expressing the antibody.

[0213] In another aspect, disclosed herein is an antibody gene comprising a variable region as recited in SEQ ID NO: 29, or a variant thereof. In one aspect, a cell line expresses the antibody.

[0214] In another aspect, the invention relates to a method for disaggregating preformed beta-amyloid fibers, comprising interacting an hC2 antibody with preformed beta-amyloid fibers.

[0215] In another aspect, the humanized antibody or a fragment thereof of the invention protects neurons from Abeta-induced degradation.

[0216] In another aspect, the invention relates to preventing Abeta-induced neuron degradation comprising treating neurons with an effective amount of the humanized antibody or a fragment thereof according to the invention.

[0217] In another aspect, the invention relates to the use of a humanized antibody or a fragment thereof according to the invention herein for the preparation of a medicament for preventing degeneration of neurons upon exposure to Abeta oligomer.

BRIEF DESCRIPTION OF FIGURES AND SEQUENCES



[0218] 

Figure 1 (Example 2): Expression Cassette of the mouse light chain variable region of the Chimeric Antibody

Figure 2 (Example 2): Expression Cassette of the mouse heavy chain variable region of the Chimeric Antibody

Figure 3 (Example 5.2): Comparison of the mouse heavy chain variable region to the closest murine germ line sequence

Figure 4 (Example 8): Activity of purified humanized C2 antibodies

Figure 5 (Example 9): Binding activity of antibodies produced by transient expression of C2 modified CDRL2 constructs in conjunction with C2 chimeric heavy chain, compared to chimeric antibody C2ChVHAF/ChVK, produced by transient transfection and purified antibody.

Figure 6 (Example 11): Results of Immunohistochemical Binding Assay with chimeric antibody AF and humanized antibody H4K1.

Figure 7 (Example 12): Functionality of mC2 on Amyloid fibers

Figure 8 (Example 12): Binding Affinity of humanized C2 in ELISA.

Figure 9 (Example 14): Conformation specific binding of mC2 to different classes of amyloid protein. Pellet preparation in the legend to this figure refers to Aβ1-42 fibers, supernatant preparation refers to amyloid monomers.

Figure 10: Humanized C2 VK sequences compared to murine sequence and human acceptor sequences DPK15 AND JK1

Figure 11: Humanized C2 VH sequences compared to murine sequence and human acceptor sequences DP54 AND JH6

Figure 12: Complete DNA and protein sequence of light chain variable region of C2 humanized antibody, C2HuVK1

Figure 13: Complete DNA and protein sequence of light chain constant region (human C Kappa) of humanized C2 antibody

Figure 14: Complete DNA and protein sequence of heavy chain constant region (human IgG4 ser228-pro) of humanized C2 antibody

Figure 15A-C (Example 15): Results of Epitope Mapping experiments

Figure 16 (Example 13): Results of aggregation assay experiments

Figure 17 (Example 13): Results of disaggregation assay experiments

Figure 18: (Example 16): Results of neuroprotection experiments with humanized antibody C2.

SEQ ID NO: 1 Amino acid sequence of C2 HuVH AF 4 humanized heavy chain variable region (CDR1)

SEQ ID NO: 2 Amino acid sequence of C2 HuVH AF 4 humanized heavy chain variable region (CDR2)

SEQ ID NO: 3 Amino acid sequence of C2 HuVH AF 4 humanized heavy chain variable region (CDR3)

SEQ ID NO: 4 Amino acid sequence of C2 HuVK 1 humanized light chain variable region (CDR1)

SEQ ID NO: 5 Amino acid sequence of C2 HuVK 1 humanized light chain variable region (CDR2)

SEQ ID NO: 6 Amino acid sequence of C2 HuVK 1 humanized light chain variable region (CDR3)

SEQ ID NO: 7 Amino acid sequence of Aβ epitope region 2

SEQ ID NO: 8 Amino acid sequence of Aβ epitope region 1

SEQ ID NO: 9 Amino acid sequence of Aβ epitope region 2 modified

SEQ ID NO: 10 Amino acid sequence of Aβ epitope region 1 modified

SEQ ID NO: 11 Amino acid sequence of Epitope region modified complete

SEQ ID NO: 12 Amino acid sequence of C2 HuVK 1 humanized light chain variable region

SEY ID NO: 13 Amino acid sequence of C2 humanized light chain

SEQ ID NO: 14 Amino acid sequence of humanized C2 light chain constant region

SEQ ID NO: 15 Amino acid sequence of C2 HuVH AF 4 humanized heavy chain variable region

SEQ ID NO: 16 Amino acid sequence of C2 humanized heavy chain

SEQ ID NO: 17: Amino acid sequence of IG GAMMA-4 CHAIN C REGION - modified

SEQ ID NO: 18: Nucleotide sequence of CDR2 of C2 HuVH AF 4 humanised heavy chain variable region

SEQ ID NO: 19: Nucleotide sequence of CDR3 of C2 HuVH AF 4 humanised heavy chain variable region

SEQ ID NO: 20: Nucleotide sequence of CDR1 of C2 HuVK 1 humanised light chain variable region

SEQ ID NO: 21: Nucleotide sequence of C2 HuVK 1 humanized light chain variable region

SEQ ID NO: 22: Nucleotide sequence of C2 humanized light chain

SEQ ID NO: 23: Nucleotide sequence of C2 humanized light chain constant region

SEQ ID NO: 24: Nucleotide sequence of C2 HuVH AF 4 humanized heavy chain variable region

SEQ ID NO: 25: Nucleotide sequence of C2 humanized heavy chain

SEQ ID NO: 26: Nucleotide sequence of C2 humanized heavy chain constant region

SEQ ID NO: 27: Amino acid sequence of Mouse C2 Light Chain Variable Region

SEQ ID NO: 28: Amino acid sequence of Mouse C2 Heavy Chain Variable Region

SEQ ID NO: 29: Nucleotide sequence of Mouse C2 Light Chain Variable Region

SEQ ID NO: 30: Nucleotide sequence of Mouse C2 Light Chain

SEQ ID NO: 31: Nucleotide sequence of Mouse C2 Heavy Chain Variable Region

SEQ ID NO: 32: Nucleotide sequence of Mouse C2 Heavy Chain


DEFINITIONS



[0219] The terms "polypeptide", "peptide", and "protein", as used herein, are interchangeable and are defined to mean a biomolecule composed of amino acids linked by a peptide bond.

[0220] The language "diseases and disorders which are caused by or associated with amyloid or amyloid-like proteins" includes, but is not limited to, diseases and disorders caused by the presence or activity of amyloid-like proteins in monomeric, fibril, or polymeric state, or any combination of the three. Such diseases and disorders include, but are not limited to, amyloidosis, endocrine tumors, and macular degeneration.

[0221] The term "amyloidosis" refers to a group of diseases and disorders associated with amyloid plaque formation including, but not limited to, secondary amyloidosis and age-related amyloidosis such as diseases including, but not limited to, neurological disorders such as Alzheimer's Disease (AD), including diseases or conditions characterized by a loss of cognitive memory capacity such as, for example, mild cognitive impairment (MCI), Lewy body dementia, Down's syndrome, hereditary cerebral hemorrhage with amyloidosis (Dutch type); the Guam Parkinson-Dementia complex; as well as other diseases which are based on or associated with amyloid-like proteins such as progressive supranuclear palsy, multiple sclerosis; Creutzfeld Jacob disease, Parkinson's disease, HIV-related dementia, ALS (amyotropic lateral sclerosis), inclusion-body myositis (IBM), Adult Onset Diabetes, and senile cardiac amyloidosis; and various eye diseases including macular degeneration, drusen-related optic neuropathy, and cataract due to beta-amyloid deposition.

[0222] The terms "detecting" or "detected" as used herein mean using known techniques for detection of biologic molecules such as immunochemical or histological methods and refer to qualitatively or quantitatively determining the presence or concentration of the biomolecule under investigation.

[0223] "Polymeric soluble amyloid" refers to multiple aggregated monomers of amyloid peptides, or of amyloid-like peptides, or of modified or truncated amyloid peptides or of other derivates of amyloid peptides forming oligomeric or polymeric structures which are soluble in the mammalian or human body more particularly in the brain, but particularly to multiple aggregated monomers of amyloid β (Aβ) or of modified or truncated amyloid β (Aβ) peptides or of derivatives thereof, which are soluble in the mammalian or human body more particularly in the brain.

[0224] "Amyloid β, Aβ or β-amyloid" is an art recognized term and refers to amyloid β proteins and peptides, amyloid β precursor protein (APP), as well as modifications, fragments and any functional equivalents thereof. In particular, by amyloid β as used herein is meant any fragment produced by proteolytic cleavage of APP but especially those fragments which are involved in or associated with the amyloid pathologies including, but not limited to, Aβ1-38, Aβ1-39, Aβ1-40, Aβ1-41 Aβ1-42 and Aβ1-43.

[0225] The structure and sequences of the amyloid β peptides as mentioned above are well known to those skilled in the art and methods of producing said peptides or of extracting them from brain and other tissues are described, for example, in Glenner and Wong, Biochem Biophys Res Comm129, 885-890 (1984). Moreover, amyloid β peptides are also commercially available in various forms.

[0226] By "isolated" is meant a biological molecule free from at least some of the components with which it naturally occurs.

[0227] The terms "antibody" or "antibodies" as used herein are art-recognized terms and are understood to refer to molecules or active fragments of molecules that bind to known antigens, particularly to immunoglobulin molecules and to immunologically active portions of immunoglobulin molecules, i.e molecules that contain a binding site that specifically binds an antigen. An immunoglobulin is a protein comprising one or more polypeptides substantially encoded by the immunoglobulin kappa and lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as myriad immunoglobulin variable region genes. Light chains are classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively. Also subclasses of the heavy chain are known. For example, IgG heavy chains in humans can be any of IgG1, IgG2, IgG3 and IgG4 subclass. The immunoglobulin according to the invention can be of any class (IgG, IgM, IgD, IgE, IgA and IgY) or subclass (IgGl, IgG2, IgG3, IgG4, IgAl and IgA2) of immunoglobulin molecule.

[0228] As used herein "specifically binds" in reference to an antibody means that the antibody binds to its target antigen with greater affinity that it does to a structurally different antigen(s).

[0229] A typical immunoglobulin structural unit is known to comprise a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25 kD) and one "heavy" chain (about 50-70 kD). The N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The terms variable light chain (VL) and variable heavy chain (VH) refer to these light and heavy chains respectively.

[0230] Antibodies exist as full length intact antibodies or as a number of well-characterized fragments produced by digestion with various peptidases or chemicals. Thus, for example, pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab')2, a dimer of Fab which itself is a light chain joined to VH-CH1 by a disulfide bond. The F(ab')2 may be reduced under mild conditions to break the disulfide linkage in the hinge region thereby converting the F(ab')2 dimer into an Fab' monomer. The Fab' monomer is essentially a Fab fragment with part of the hinge region (see, Fundamental Immunology, W. E. Paul, ed., Raven Press, N.Y. (1993), for a more detailed description of other antibody fragments). While various antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that any of a variety of antibody fragments may be synthesized de novo either chemically or by utilizing recombinant DNA methodology. Thus, the term antibody, as used herein also includes antibody fragments either produced by the modification of whole antibodies or synthesized de novo or antibodies and fragments obtained by using recombinant DNA methodologies.

[0231] "Antibodies" are intended within the scope of the present invention to include monoclonal antibodies, polyclonal antibodies, chimeric, single chain, bispecific, simianized, human and humanized antibodies as well as active fragments thereof. Examples of active fragments of molecules that bind to known antigens include separated light and heavy chains, Fab, Fab/c, Fv, Fab', and F(ab')2 fragments, including the products of an Fab immunoglobulin expression library and epitope-binding fragments of any of the antibodies and fragments mentioned above.

[0232] These active fragments can be derived from an antibody of the present invention by a number of techniques. For example, monoclonal antibodies can be cleaved with an enzyme, such as pepsin, and subjected to HPLC gel filtration. The appropriate fraction containing Fab fragments can then be collected and concentrated by membrane filtration and the like. For further description of general techniques for the isolation of active fragments of antibodies, see for example, Khaw, B. A. et al. J. Nucl. Med. 23:1011-1019 (1982); Rousseaux et al. Methods Enzymology, 121:663-69, Academic Press, 1986.

[0233] Recombinantly made antibodies may be conventional full length antibodies, active antibody fragments known from proteolytic digestion, unique active antibody fragments such as Fv or single chain Fv (scFv), domain deleted antibodies, and the like. An Fv antibody is about 50 Kd in size and comprises the variable regions of the light and heavy chain. A single chain Fv ("scFv") polypeptide is a covalently linked VH::VL heterodimer which may be expressed from a nucleic acid including VH- and VL-encoding sequences either joined directly or joined by a peptide-encoding linker. See Huston, et al. (1988) Proc. Nat. Acad. Sci. USA, 85:5879-5883. A number of structures for converting the naturally aggregated, but chemically separated light and heavy polypeptide chains from an antibody V region into an scFv molecule which will fold into a three dimensional structure substantially similar to the structure of an antigen-binding site. See, e.g. U.S. Patent Nos. 5,091,513, 5,132,405 and 4,956,778.

[0234] The combining site refers to the part of an antibody molecule that participates in antigen binding. The antigen binding site is formed by amino acid residues of the N-terminal variable ("V") regions of the heavy ("H") and light ("L") chains. The antibody variable regions comprise three highly divergent stretches referred to as "hypervariable regions" or "complementarity determining regions" (CDRs) which are interposed between more conserved flanking stretches known as "framework regions" (FRs). In an antibody molecule, the three hypervariable regions of a light chain (LCDR1, LCDR2, and LCDR3) and the three hypervariable regions of a heavy chain (HCDR1, HCDR2 and HCDR3) are disposed relative to each other in three dimensional space to form an antigen binding surface or pocket. The antibody combining site therefore represents the amino acids that make up the CDRs of an antibody and any framework residues that make up the binding site pocket.

[0235] The identity of the amino acid residues in a particular antibody that make up the combining site can be determined using methods well known in the art. For example, antibody CDRs may be identified as the hypervariable regions originally defined by Kabat et al. (see, "Sequences of Proteins of Immunological Interest," E. Kabat et al., U.S. Department of Health and Human Services; Johnson, G and Wu, TT (2001) Kabat Database and its applications: future directions. Nucleic Acids Research, 29: 205-206; http://immuno.bme.nwa.edu). The positions of the CDRs may also be identified as the structural loop structures originally described by Chothia and others, (see Chothia and Lesk, J. Mol. Biol. 196, 901 (1987), Chothia et al., Nature 342, 877 (1989), and Tramontano et al., J. Mol. Biol. 215, 175 (1990)). Other methods include the "AbM definition" which is a compromise between Kabat and Chothia and is derived using Oxford Molecular's AbM antibody modeling software (now Accelrys) or the "contact definition" of CDRs by Macallum et al., ("Antibody-antigen interactions: contact analysis and binding site topography," J Mol Biol. 1996 Oct 11;262(5):732-45). The following chart identifies CDRs based upon various known definitions.
Loop Kabat AbM Chothia Contact
L1 L24 -- L34 L24 -- L34 L24 -- L34 L30 -- L36
L2 L50 -- L56 L50 -- L56 L50 -- L56 L46 -- L55
L3 L89 -- L97 L89 -- L97 L89 -- L97 L89 -- L96
H1 H31 -- H35B H26 -- H35B H26 -- H32..34 H30 -- H35B
(Kabat Numbering)
H1 H31 -- H35 H26 -- H35 H26 -- H32 H30 -- H35
(Chothia Numbering)
H2 H50 -- H65 H50 -- H58 H52 -- H56 H47 -- H58
H3 H95 -- H102 H95 -- H102 H95 -- H102 H93 -- H101
General guidelines by which one may identify the CDRs in an antibody from sequence alone are as follows:

LCDR1:

Start - Approximately residue 24.

Residue before is always a Cys.

Residue after is always a Trp. Typically TRP is followed with TYR-GLN, but also may be followed by LEU-GLN, PHE-GLN, or TYR-LEU.

Length is 10 to 17 residues.

LCDR2:

Start - 16 residues after the end of L1.

Sequence before is generally ILE-TYR, but also may be VAL-TYR, ILE-LYS, or ILE-PHE. Length is generally 7 residues.

LCDR3:

Start - generally 33 residues after end of L2.

Residue before is a Cys.

Sequence after is PHE-GLY-X-GLY.

Length is 7 to 11 residues.

HCDR1:

Start - at approximately residue 26 (four residues after a CYS) [Chothia / AbM definition] Kabat definition starts 5 residues later.

Sequence before is CYS-X-X-X.

Residues after is a TRP, typically followed by VAL, but also followed by ILE, or ALA. Length is 10 to 12 residues under AbM definition while Chothia definition excludes the last 4 residues.

HCDR2:

Start - 15 residues after the end of Kabat /AbM definition of CDR-H1.

Sequence before typically LEU-GLU-TRP-ILE-GLY, but a number of variations are possible.

Sequence after is LYS/ARG-LEU/ILE/VAL/PHE/THR/ALA-THR/SER/ILE/ALA

Length is 16 to 19 residues under Kabat definition (AbM definition ends 7 residues earlier).

HCDR3:

Start -33 residues after end of CDR-H2 (two residues after a CYS).

Sequence before is CYS-X-X (typically CYS-ALA-ARG).

Sequence after is TRP-GLY-X-GLY.

Length is 3 to 25 residues.



[0236] The identity of the amino acid residues in a particular antibody that are outside the CDRs, but nonetheless make up part of the combining site by having a side chain that is part of the lining of the combining site (i.e., it is available to linkage through the combining site), can be determined using methods well known in the art such as molecular modeling and X-ray crystallography. See e.g., Riechmann et al., (1988) Nature, 332:;323-327.

[0237] Chimeric antibodies are those in which one or more regions of the antibody are from one species of animal and one or more regions of the antibody are from a different species of animal. A preferred chimeric antibody is one which includes regions from a primate immunoglobulin. A chimeric antibody for human clinical use is typically understood to have variable regions from a non-human animal, e.g. a rodent, with the constant regions from a human. In contrast, a humanized antibody uses CDRs from the non-human antibody with most or all of the variable framework regions from and all the constant regions from a human immunoglobulin. A human chimeric antibody is typically understood to have the variable regions from a rodent. A typical human chimeric antibody has human heavy constant regions and human light chain constant regions with the variable regions of both the heavy and light coming from a rodent antibody. A chimeric antibody may include some changes to a native amino acid sequence of the human constant regions and the native rodent variable region sequence. Chimeric and humanized antibodies may be prepared by methods well known in the art including CDR grafting approaches (see, e.g., U.S. Patent Nos. 5,843,708; 6,180,370; 5,693,762; 5,585,089; 5,530,101), chain shuffling strategies (see e.g., U.S. Patent No. 5,565,332; Rader et al., Proc. Natl. Acad. Sci. USA (1998) 95:8910-8915), molecular modeling strategies (U.S. Patent No. 5,639,641), and the like.

[0238] A "humanized antibody" as used herein in the case of a two chain antibody is one where at least one chain is humanized. A humanized antibody chain has a variable region where one or more of the framework regions are human. A humanized antibody which is a single chain is one where the chain has a variable region where one or more of the framework regions are human. The non-human portions of the variable region of the humanized antibody chain or fragment thereof is derived from a non-human source, particularly a non-human antibody, typically of rodent origin. The non-human contribution to the humanized antibody is typically provided in form at least one CDR region which is interspersed among framework regions derived from one (or more) human immunoglobulin(s). In addition, framework support residues may be altered to preserve binding affinity.

[0239] The humanized antibody may further comprise constant regions (e.g., at least one constant region or portion thereof, in the case of a light chain, and preferably three constant regions in the case of a heavy chain). The constant regions of a humanized antibody if present generally are human.

[0240] Methods to obtain "humanized antibodies" are well known to those skilled in the art. (see, e.g., Queen et al., Proc. Natl Acad Sci USA, 86:10029-10032 (1989), Hodgson et al., Bio/Technology, 9:421 (1991)).

[0241] A "humanized antibody" may also be obtained by a novel genetic engineering approach that enables production of affinity-matured human-like polyclonal antibodies in large animals such as, for example, rabbits and mice. See, e.g. U.S. Pat No. 6,632,976.
The term constant region (CR) as used herein refers to constant regions genes of the immunoglobulin. The constant region genes encode the portion of the antibody molecule which confers effector functions. For Chimeric human antibodies and humanized antibodies, typically non-human (e.g., murine), constant regions are substituted by human constant regions. The constant regions of the subject chimeric or humanized antibodies are typically derived from human immunoglobulins. The heavy chain constant region can be selected from any of the five isotypes: alpha, delta, epsilon, gamma or mu. Further, heavy chains of various subclasses (such as the IgG subclasses of heavy chains) are responsible for different effector functions and thus, by choosing the desired heavy chain constant region, antibodies with desired effector function can be produced. Constant regions that may be used within the scope of this invention are gamma 1 (IgG1), particularly an Fc region of the gamma 1 (IgG1) isotype, gamma 3 (IgG3) and especially gamma 4 (IgG4). The light chain constant region can be of the kappa or lambda type, preferably of the kappa type. In one embodiment the light chain constant region is the human kappa constant chain (Heiter et al. (1980) Cell 22:197-207) and the heavy constant chain is the human IgG4 constant chain.

[0242] The term "monoclonal antibody" is also well recognized in the art and refers to an antibody that is the product of a single cloned antibody producing cell. Monoclonal antibodies are typically made by fusing a normally short-lived, antibody-producing B cell to a fast-growing cell, such as a cancer cell (sometimes referred to as an "immortal" cell). The resulting hybrid cell, or hybridoma, multiplies rapidly, creating a clone that produces the antibody.

[0243] For the purpose of the present invention, "monoclonal antibody" is also to be understood to comprise antibodies that are produced by a mother clone which has not yet reached full monoclonality.

[0244] "Functionally equivalent antibody" is understood to refer to an antibody which substantially shares at least one major functional property with an antibody mentioned above and herein described comprising: binding specificity to the β-amyloid protein, particularly to the Aβ1-42 protein, and more particularly to the 16-21 epitope region of the Aβ1-42 protein, immunoreactivity in vitro, inhibition of aggregation of the Aβ1-42 monomers into high molecular polymeric fibrils and/or disaggregation of preformed Aβ1-42 polymeric fibrils, and/or a β-sheet breaking property and alleviating the effects of amyloidosis, a group of diseases and disorders associated with amyloid plaque formation including secondary amyloidosis and age-related amyloidosis such as diseases including, but not limited to, neurological disorders such as Alzheimer's Disease (AD), Lewy body dementia, Down's syndrome, hereditary cerebral hemorrhage with amyloidosis (Dutch type); the Guam Parkinson-Dementia complex; as well as other diseases which are based on or associated with amyloid-like proteins such as progressive supranuclear palsy, multiple sclerosis; Creutzfeld Jacob disease, Parkinson's disease, HIV-related dementia, ALS (amyotropic lateral sclerosis), Adult Onset Diabetes; senile cardiac amyloidosis; endocrine tumors, and others, including macular degeneration, when administered prophylactically or therapeutically. The antibodies can be of any class such as IgG, IgM, or IgA, etc or any subclass such as IgG1, IgG2a, etc and other subclasses mentioned herein above or known in the art, but particularly of the IgG4 class. Further, the antibodies can be produced by any method, such as phage display, or produced in any organism or cell line, including bacteria, insect, mammal or other type of cell or cell line which produces antibodies with desired characteristics, such as humanized antibodies. The antibodies can also be formed by combining a Fab portion and an Fc region from different species.

[0245] The term "hybridize" as used refers to conventional hybridization conditions, preferably to hybridization conditions at which 5xSSPE, 1% SDS, 1xDenhardts solution is used as a solution and/or hybridization temperatures are between 35°C and 70°C, preferably 65°C. After hybridization, washing is preferably carried out first with 2xSSC, 1% SDS and subsequently with 0.2xSSC at temperatures between 35°C and 70°C, preferably at 65°C (regarding the definition of SSPE, SSC and Denhardts solution see Sambrook et al. loc. cit.). Stringent hybridization conditions as for instance described in Sambrook et al, supra, are particularly preferred. Particularly preferred stringent hybridization conditions are for instance present if hybridization and washing occur at 65°C as indicated above. Non-stringent hybridization conditions, for instance with hybridization and washing carried out at 45°C are less preferred and at 35°C even less.

[0246] "Homology" between two sequences is determined by sequence identity. If two sequences which are to be compared with each other differ in length, sequence identity preferably relates to the percentage of the nucleotide residues of the shorter sequence which are identical with the nucleotide residues of the longer sequence. Sequence identity can be determined conventionally with the use of computer programs such as the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive Madison, WI 53711). Bestfit utilizes the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2 (1981), 482-489, in order to find the segment having the highest sequence identity between two sequences. When using Bestfit or another sequence alignment program to determine whether a particular sequence has for instance 95% identity with a reference sequence of the present invention, the parameters are preferably so adjusted that the percentage of identity is calculated over the entire length of the reference sequence and that homology gaps of up to 5% of the total number of the nucleotides in the reference sequence are permitted. When using Bestfit, the so-called optional parameters are preferably left at their preset ("default") values. The deviations appearing in the comparison between a given sequence and the above-described sequences of the invention may be caused for instance by addition, deletion, substitution, insertion or recombination. Such a sequence comparison can preferably also be carried out with the program "fasta20u66" (version 2.0u66, September 1998 by William R. Pearson and the University of Virginia; see also W.R. Pearson (1990), Methods in Enzymology 183, 63-98, appended examples and http://workbench.sdsc.edu/). For this purpose, the "default" parameter settings maybe used.

[0247] The antibody according to the invention may be an immunoglobulin or antibody, which is understood to have each of its binding sites identical (if multivalent) or, in the alternative, may be a "bispecific" or "bifunctional antibody".

[0248] A "bispecific" or "bifunctional antibody" is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites. Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann, Clin. Exp. Immunol. 79:315-321 (1990); Kostelny et al., J. Immunol. 148, 1547-1553 (1992).

[0249] The term "fragment" refers to a part or portion of an antibody or antibody chain comprising fewer amino acid residues than an intact or complete antibody or antibody chain. Fragments can be obtained via chemical or enzymatic treatment of an intact or complete antibody or antibody chain. Fragments can also be obtained by recombinant means. Exemplary fragments include Fab, Fab', F(ab')2, Fabc and/or Fv fragments. The term "antigen-binding fragment" refers to a polypeptide fragment of an immunoglobulin or antibody that binds antigen or competes with intact antibody (i.e., with the intact antibody from which they were derived) for antigen binding (i.e., specific binding).

[0250] Binding fragments are produced by recombinant DNA techniques, or by enzymatic or chemical cleavage of intact immunoglobulins. Binding fragments include Fab, Fab', F(ab')2, Fabc, Fv, single chains, and single-chain antibodies.

[0251] "Fragment" also refers to a peptide or polypeptide comprising an amino acid sequence of at least 5 contiguous amino acid residues, at least 10 contiguous amino acid residues, at least 15 contiguous amino acid residues, at least 20 contiguous amino acid residues, at least 25 contiguous amino acid residues, at least 40 contiguous amino acid residues, at least 50 contiguous amino acid residues, at least 60 contiguous amino residues, at least 70 contiguous amino acid residues, at least contiguous 80 amino acid residues, at least contiguous 90 amino acid residues, at least contiguous 100 amino acid residues, at least contiguous 125 amino acid residues, at least 150 contiguous amino acid residues, at least contiguous 175 amino acid residues, at least contiguous 200 amino acid residues, or at least contiguous 250 amino acid residues of the amino acid sequence of another polypeptide. In a specific embodiment, a fragment of a polypeptide retains at least one function of the polypeptide.

[0252] The term "antigen" refers to an entity or fragment thereof which can bind to an antibody. An immunogen refers to an antigen which can elicit an immune response in an organism, particularly an animal, more particularly a mammal including a human. The term antigen includes regions known as antigenic determinants or epitopes which refers to a portion of the antigen (which are contacted or which play a significant role in supporting a contact reside in the antigen responsible for antigenicity or antigenic determinants.

[0253] As used herein, the term "soluble" means partially or completely dissolved in an aqueous solution.

[0254] Also as used herein, the term "immunogenic" refers to substances which elicit the production of antibodies, T-cells and other reactive immune cells directed against an antigen of the immunogen.

[0255] An immune response occurs when an individual produces sufficient antibodies, T-cells and other reactive immune cells against administered immunogenic compositions of the present invention to moderate or alleviate the disorder to be treated.

[0256] The term immunogenicity as used herein refers to a measure of the ability of an antigen to elicit an immune response (humoral or cellular) when administered to a recipient. The present invention is concerned with approaches that reduce the immunogenicity of the subject human chimeric or humanized antibodies.

[0257] Humanized antibody of reduced immunogenicity refers to a humanized antibody exhibiting reduced immunogenicity relative to the parent antibody, e.g., the murine antibody.

[0258] Humanized antibody substantially retaining the binding properties of the parent antibody refers to a humanized antibody which retains the ability to specifically bind the antigen recognized by the parent antibody used to produce such humanized antibody. Preferably the humanized antibody will exhibit the same or substantially the same antigen-binding affinity and avidity as the parent antibody. Ideally, the affinity of the antibody will not be less than 10% of the parent antibody affinity, more preferably not less than about 30%, and most preferably the affinity will not be less than 50% of the parent antibody. Methods for assaying antigen-binding affinity are well known in the art and include half-maximal binding assays, competition assays, and Scatchard analysis. Suitable antigen binding assays are described in this application.

[0259] A "back mutation" is a mutation introduced in a nucleotide sequence which encodes a humanized antibody, the mutation results in an amino acid corresponding to an amino acid in the parent antibody (e.g., donor antibody, for example, a murine antibody). Certain framework residues from the parent antibody may be retained during the humanization of the antibodies of the invention in order to substantially retain the binding properties of the parent antibody, while at the same time minimizing the potential immunogenicity of the resultant antibody. In one embodiment of the invention, the parent antibody is of mouse origin. For example, the back mutation changes a human framework residue to a parent murine residue. Examples of framework residues that may be back mutated include, but are not limited to, canonical residues, interface packing residues, unusual parent residues which are close to the binding site, residues in the "Vernier Zone" (which forms a platform on which the CDRs rest) (Foote & Winter, 1992, J. Mol. Biol. 224, 487-499), and those close to CDR H3.

[0260] As used herein a "conservative change" refers to alterations that are substantially conformationally or antigenically neutral, producing minimal changes in the tertiary structure of the mutant polypeptides, or producing minimal changes in the antigenic determinants of the mutant polypeptides, respectively, as compared to the native protein. When referring to the antibodies and antibody fragments of the invention, a conservative change means an amino acid substitution that does not render the antibody incapable of binding to the subject receptor. Those of ordinary skill in the art will be able to predict which amino acid substitutions can be made while maintaining a high probability of being conformationally and antigenically neutral. Such guidance is provided, for example in Berzofsky, (1985) Science 229:932-940 and Bowie et al. (1990) Science 247:1306-1310. Factors to be considered that affect the probability of maintaining conformational and antigenic neutrality include, but are not limited to: (a) substitution of hydrophobic amino acids is less likely to affect antigenicity because hydrophobic residues are more likely to be located in a protein's interior; (b) substitution of physiochemically similar, amino acids is less likely to affect conformation because the substituted amino acid structurally mimics the native amino acid; and (c) alteration of evolutionarily conserved sequences is likely to adversely affect conformation as such conservation suggests that the amino acid sequences may have functional importance. One of ordinary skill in the art will be able to assess alterations in protein conformation using well-known assays, such as, but not limited to microcomplement fixation methods (Wasserman et al. (1961) J. Immunol. 87:290-295; Levine et al. (1967) Meth. Enzymol. 11:928-936) and through binding studies using conformation-dependent monoclonal antibodies (Lewis et al. (1983) Biochem. 22:948-954).

[0261] Further, the term "therapeutically effective amount" refers to the amount of antibody which, when administered to a human or animal, which is sufficient to result in a therapeutic effect in said human or animal. The effective amount is readily determined by one of skill in the art following routine procedures.

[0262] As used herein, the terms "treat," "prevent," "preventing," and "prevention" refer to the prevention of the recurrence or onset of one or more symptoms of a disorder in a subject resulting from the administration of a prophylactic or therapeutic agent.

Construction of Humanized Antibodies



[0263] The present invention provides novel uses and compositions comprising highly specific and highly effective antibodies having the ability to specifically recognize and bind to specific epitopes from a range of β-amyloid antigens. The antibodies enabled by the teaching of the present invention are particularly useful for the treatment of amyloidosis, a group of diseases and disorders associated with amyloid plaque formation including secondary amyloidosis and age-related amyloidosis including, but not limited to, neurological disorders such as Alzheimer's Disease (AD), Lewy body dementia, Down's syndrome, hereditary cerebral hemorrhage with amyloidosis (Dutch type); the Guam Parkinson-Dementia complex; as well as other diseases which are based on or associated with amyloid-like proteins such as progressive supranuclear palsy, multiple sclerosis; Creutzfeld Jacob disease, hereditary cerebral hemorrhage with amyloidosis Dutch type, Parkinson's disease, HIV-related dementia, ALS (amyotropic lateral sclerosis), Adult Onset Diabetes; senile cardiac amyloidosis; endocrine tumors, and others, including macular degeneration, to name just a few.

[0264] A fully humanized or reshaped variable region according to the present invention may be created within the scope of the invention by first designing a variable region amino acid sequence that contains CDRs derived from murine antibody ACI-01-Ab7C2 (named "mC2" throughout the application and deposited 01 December 2005 with the "Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) in Braunschweig, Mascheroder Weg 1 B, 38124 Branuschweig, under the provisions of the Budapest Treaty and given accession no DSM ACC2750) embedded in human-derived framework sequences. The non-human-, particularly the rodent-derived CDRs, which may be obtained from the antibody according to the present invention, provide the desired specificity. Accordingly, these residues are to be included in the design of the reshaped variable region essentially unchanged. Any modifications should thus be restricted to a minimum and closely watched for changes in the specificity and affinity of the antibody. On the other hand, framework residues in theory can be derived from any human variable region.

[0265] In order to create a reshaped antibody which shows an acceptable or an even improved affinity, a human framework sequences should be chosen, which is equally suitable for creating a reshaped variable region and for retaining antibody affinity.

[0266] In order to achieve this goal, the best-fit strategy was developed. As it is known that the framework sequences serve to hold the CDRs in their correct spatial orientation for interaction with antigen, and that framework residues can sometimes even participate in antigen binding, this strategy aims at minimizing changes that may negatively effect the three-dimensional structure of the antibody by deriving the human framework sequence used for antibody reshaping from the human variable region that is most homologous or similar to the non-human-, particularly the rodent-derived variable region. This will also maximise the likelihood that affinity will be retained in the reshaped antibody.

[0267] At its simplest level, the "best fit" strategy involves comparing the donor rodent V-region with all known human V-region amino acid sequences, and then selecting the most homologous to provide the acceptor framework regions for the humanization exercises. In reality there are several other factors which should be considered, and which may influence the final selection of acceptor framework regions. Molecular modelling predictions may be used in this regard prior to any experimental work in an attempt to maximise the affinity of the resultant reshaped antibody. Essentially, the goal of the modelling is to predict which key residues (if any) of the most homologous human framework should be left as in the rodent to obtain the best affinity in the reshaped antibody.

[0268] In one embodiment of the invention, the CDRs are obtainable from mouse monoclonal antibody, particularly from mouse monoclonal antibody ACI-01-Ab7C2 (named "mC2" throughout the application) described in co-pending application EP 05 02 7092.5 filed 12.12.2005.

[0269] Hybridoma cells FP-12H3-C2, producing mouse monoclonal antibody ACI-01-Ab7C2 (named "mC2" and hC2 for the humanized C2 antibody, throughout the application) were deposited 01 December 2005 in co-pending application no EP05027092.5 with the "Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) in Braunschweig, Mascheroder Weg 1 B, 38124 Braunschweig, under the provisions of the Budapest Treaty and given accession no DSM ACC2750.

[0270] The mouse antibody may be raised against a supramolecular antigenic construct comprising an antigenic peptide corresponding to the amino acid sequence of the β-amyloid peptide, particularly of β-amyloid peptide Aβ1-15, Aβ1-16 and Aβ1-16(Δ14), modified with a hydrophobic moiety such as, for example, palmitic acid or a hydrophilic moiety such as, for example, polyethylene glycol (PEG) or a combination of both, wherein the hydrophobic and hydrophilic moiety, respectively, is covalently bound to each of the termini of the antigenic peptide through at least one, particularly one or two amino acids such as, for example, lysine, glutamic acid and cysteine or any other suitable amino acid or amino acid analogue capable of serving as a connecting device for coupling the hydrophobic and hydrophilic moiety to the peptide fragment. When a PEG is used as the hydrophilic moiety, the free PEG termini is covalently bound to phosphatidylethanolamine or any other compound suitable to function as the anchoring element, for example, to embed the antigenic construct in the bilayer of a liposome.

[0271] In particular, a mouse antibody may be raised against a supramolecular antigenic construct comprising an antigenic peptide corresponding to the amino acid sequence of the β-amyloid peptide Aβ1-16 modified with a hydrophilic moiety such as, for example, polyethylene glycol (PEG) hydrophilic moiety is covalently bound to each of the termini of the antigenic peptide through at least one, particularly one or two amino acids such as, for example, lysine, glutamic acid and cysteine or any other suitable amino acid or amino acid analogue capable of serving as a connecting device for coupling the hydrophobic and hydrophilic moiety to the peptide fragment. When a PEG is used as the hydrophilic moiety, the free PEG termini are covalently bound to phosphatidylethanolamine or any other compound suitable to function as the anchoring element, for example, to embed the antigenic construct in the bilayer of a liposome.

[0272] In an embodiment of the invention, a humanized antibody or a fragment thereof is provided which comprises in the variable region at least one CDR of non-human origin embedded in one or more human- or primate-derived framework regions and combined with a constant region derived from a human or primate source antibody, which chimeric antibody or a fragment thereof, or a humanized antibody or a fragment thereof is capable of specifically recognizing and binding β-amyloid monomeric peptide.

[0273] The CDRs contain the residues most likely to bind antigen and must be retained in the reshaped antibody. CDRs are defined by sequence according to Kabat et al., Sequence of Proteins of Immunological Interest, 5th Edition, The United States Department of Health and Human Services, The United States Government Printing Office, 1991. CDRs fall into canonical classes (Chothia et al, 1989 Nature, 342, 877-883) where key residues determine to a large extent the structural conformation of the CDR loop. These residues are almost always retained in the reshaped antibody.

[0274] In the process for preparing a humanized antibody according to the invention, the amino acid sequences of the C2 heavy chain and light chain variable regions (VH and VK) are compared to rodent antibody VH and VK sequences in the NCBI and Kabat databases.

[0275] The closest match mouse germ line gene to C2 VK is bbl, Locus MMU231201, (Schable et al, 1999). A comparison reveals that two amino acids differ from this germ line sequence, both located within CDRL1. Mature murine antibodies with similar, but not identical, sequence can be found. Several have an identical CDRL2 and identical CDRL3, but the CDRL1 of C2 seems to be unique. Comparison with human germ line VK sequences shows that genes from subgroup VKII are the best match for C2 VK (Cox et al, 1994). C2 VK can thus be assigned to Kabat subgroup MuVKII.Sequence.

[0276] DPK15 together with the human J region HuJK1 may be selected to provide the acceptor framework sequences for the humanized VK.

[0277] The residues at the interface between the variable light and heavy chains have been defined (Chothia et al, 1985 J. Mol. Biol., 186, 651-663). These are usually retained in the reshaped antibody. The Phe at position 87 of mouse C2 VK is unusual at the interface, where a Tyr is more common in the VKII subgroup, indicating that this framework residue may be important for antibody activity. Tyr 87 is present in the human germline and humanized C2VK.

[0278] The humanized VK sequences thus may be designed such that the C2HuVK1 consists of mouse C2 VK CDRs with frameworks from DPK 15 and human JK1. In a specific embodiment of the invention, murine residues may be substituted in the human framework region at positions 45, and/or 87. In the CDR2 region obtainable from a mouse monoclonal antibody, particularly murine antibody ACI-01-Ab7C2, amino acid substitutions may be made at Kabat positions 50 and/or 53. Residue 45 may be involved in supporting the conformation of the CDRs. Residue 87 is located at the interface of the VH and VK domains. Therefore these residues may be critical for maintenance of antibody binding.

[0279] The closest match mouse germ line gene to C2 VH AF is VH7183, Locus AF120466, (Langdon et al, 2000). Comparison with human germ line VH sequences shows that genes from subgroup VHIII are the best match for C2 VH. C2 VH AF can be assigned to Kabat subgroup MuVHIIID. Sequence DP54 together with the human J region HuJH6 can be selected to provide the acceptor framework sequences for the humanized VH.
The comparison shows that there are nine amino acid differences between the C2 VH sequences and the human acceptor germ line sequence DP54 and JH6, most being located within CDRH2. Mature murine antibodies with identical or similar (one residue different) CDRH1 or with similar CDRH2 (one residue different) are found, but none with all three CDRs identical to C2 VH AF. CDRH3 of C2 antibody is unusually short, consisting of only three residues. However, other antibodies are found in the database with CDRH3 of this length. Residue 47 of C2 VH is Leu rather than the more common Trp, and residue 94 is Ser rather than the normal Arg, indicating that these framework residues may be important for antibody activity.

[0280] Various humanized VH sequences may be designed. C2HuVH1 consists of C2 VH AF CDRs with frameworks from DP54 and HuJH6. In a specific embodiment of the invention, murine residues may be substituted in the human framework region at positions 47 or 94 or both. Residue 47 in framework 2 makes contact both with the CDRs and with the VK domain. Residue 94 may be involved in supporting the conformation of the CDRs. Therefore these residues may be critical for maintenance of antibody binding.

[0281] Different HCVR and LCVR regions may be designed which comprise the non-human CDRs obtainable from the donor antibody, for example, a murine antibody, embedded into the native or modified human- or primate-derived framework regions. The modification may particularly concern an exchange of one or more amino acid residues within the framework region by non-human residues, particularly murine residues, more commonly found in this position in the respective subgroups or by residues which have similar properties to the ones more commonly found in this position in the respective subgroups.

[0282] The modification of the framework region the framework sequences serve to hold the CDRs in their correct spatial orientation for interaction with antigen, and that framework residues can sometimes even participate in antigen binding. In one embodiment of the invention measures are taken to further adapt the selected human framework sequences to make them most similar to the sequences of the rodent frameworks in order to maximise the likelihood that affinity will be retained in the reshaped antibody.

[0283] Accordingly, murine residues in the human framework region may be substituted. In particular, murine residues may be substituted in the human framework region of the Heavy Chain Variable (HCVR) region at positions 47 or 94 or both and in the human framework region of the Light Chain Variable (LCVR) region at positions 45 and/or 87. In the CDR2 region obtainable from a mouse monoclonal antibody, particularly murine antibody ACI-01-Ab7C2, amino acid substitutions may be made at Kabat positions 50 and/or 53..

[0284] The residues found in the above indicated positions in the human framework region may be exchanged by murine residues more commonly found in this position in the respective subgroups. In particular, the Trp in Kabat position 47 in the human- or primate-derived framework region of the Heavy Chain Variable Region as shown in SEQ ID NO: 15 may be replaced by an Leu or by an amino acid residue that has similar properties and the substitution of which leads to alterations that are substantially conformationally or antigenically neutral, producing minimal changes in the tertiary structure of the mutant polypeptides, or producing minimal changes in the antigenic determinants. In particular, the Trp in Kabat position 47 in the human- or primate-derived framework region of the Heavy Chain Variable Region as shown in SEQ ID NO: 15 may further be replaced by an amino acid selected from the group consisting of norleucine, Ile, Val, Met, Ala, and Phe, particularly by Ile. Alternative conservative substitutions may be contemplated which are conformationally and antigenically neutral.
The Arg in Kabat position 94 in the human- or primate-derived framework region of the Heavy Chain Variable Region as shown in SEQ ID NO: 15 may be replaced by Ser or by an amino acid residue that has similar properties and the substitution of which leads to alterations that are substantially conformationally or antigenically neutral, producing minimal changes in the tertiary structure of the mutant polypeptides, or producing minimal changes in the antigenic determinants. In particular, the Arg in Kabat position 94 in the human- or primate-derived framework region of the Heavy Chain Variable Region as shown in SEQ ID NO: 15 may alternatively be replaced by Thr.

[0285] In another alternative, both residues may be replaced in the humanized antibody.
The Gln in Kabat position 45 in the human- or primate-derived framework region of the Light Chain Variable Region as shown in SEQ ID NO: 12 may be replaced by Lys or by an amino acid residue that has similar properties and the substitution of which leads to alterations that are substantially conformationally or antigenically neutral, producing minimal changes in the tertiary structure of the mutant polypeptides, or producing minimal changes in the antigenic determinants. In particular, the Gln in Kabat position 45 in the human- or primate-derived framework region of the Light Chain Variable Region as shown in SEQ ID NO: 12 may be replaced by an amino acid selected from the group consisting of Arg, Gln, and Asn, particularly by Arg.

[0286] The Leu in Kabat position 50 in the human- or primate-derived framework region of the Light Chain Variable Region as shown in SEQ ID NO: 12 may be replaced by Lys or by an amino acid residue that has similar properties and the substitution of which leads to alterations that are substantially conformationally or antigenically neutral, producing minimal changes in the tertiary structure of the mutant polypeptides, or producing minimal changes in the antigenic determinants. In particular, the Leu in Kabat position 50 in the human- or primate-derived framework region of the Light Chain Variable Region as shown in SEQ ID NO: 12 may be replaced by an amino acid selected from the group consisting of Arg, Gln, and Asn, particularly by Arg.

[0287] The Asn in Kabat position 53 in the human- or primate-derived framework region of the Light Chain Variable Region as shown in SEQ ID NO: 12 may be replaced by His and Gln or by an amino acid residue that has similar properties and the substitution of which leads to alterations that are substantially conformationally or antigenically neutral, producing minimal changes in the tertiary structure of the mutant polypeptides, or producing minimal changes in the antigenic determinants. In particular, the Asn in Kabat position 53 in the human- or primate-derived framework region of the Light Chain Variable Region as shown in SEQ ID NO: 12 may be replaced by an amino acid selected from the group consisting of Gln, His, Lys and Arg.

[0288] The Thr in Kabat position 87 in the human- or primate-derived framework region of the Light Chain Variable Region as shown in SEQ ID NO: 12 may be replaced by Phe or by an amino acid residue that has similar properties and the substitution of which leads to alterations that are substantially conformationally or antigenically neutral, producing minimal changes in the tertiary structure of the mutant polypeptides, or producing minimal changes in the antigenic determinants. In particular, the Tyr in Kabat position 87 in the human- or primate-derived framework region of the Light Chain Variable Region as shown in SEQ ID NO: 12 may be replaced by an amino acid selected from the group consisting of Leu, Val, Ile, and Ala, particularly by Leu.

[0289] The so obtained variable region comprising at least one CDR of non-human origin embedded in one or more human- or primate-derived framework regions may then be combined with a constant region derived from a human or primate source antibody, particularly with human IgG4 or κ constant regions respectively. The IgG4 constant region may be modified by, for example, changing Serine at position 228 in the hinge region to Proline (HuIgG4 Ser-Pro). This mutation stabilizes the interchain disulphide bond and prevents the formation of half molecules that may occur in native human IgG4 preparations. The IgG4 constant region may be further modified by deletion of the terminal Lys in position 439 as shown in SEQ ID NO: 16.

[0290] The modified variable regions may be constructed by method known in the art such as, for example overlapping PCR recombination. The expression cassettes for the chimeric antibody, C2 ChVH AF and C2 ChVK, may be used as templates for mutagenesis of the framework regions to the required sequences. Sets of mutagenic primer pairs are synthesized encompassing the regions to be altered. The humanized VH and VK expression cassettes produced may be cloned into appropriate cloning vectors know in the art such as, for example, pUC19. After the entire DNA sequence is confirmed to be correct for each VH and VK, the modified heavy and light chain V-region genes can be excised from the cloning vector as expression cassettes. These can then be transferred to appropriate expression vectors such as pSVgpt and pSVhyg which include human IgG4 Ser-Pro or κ constant regions respectively.

EXPRESSION VECTORS



[0291] Expression vector pSVgpt is based on pSV2gpt (Mulligan and Berg, 1980) and includes the ampicillin resistance gene for selection in bacterial cells, the gpt gene for selection in mammalian cells, the murine heavy chain immunoglobulin enhancer region, genomic sequence encoding the constant region gene and SV40 poly A sequences. The heavy chain variable region for expression is inserted as a HindIII to BamHI fragment.

[0292] Expression vector pSVhyg includes the ampicillin resistance gene for selection in bacterial cells, the hyg gene for selection in mammalian cells, the murine heavy chain immunoglobulin enhancer region, genomic sequence encoding the kappa constant region gene and including the kappa enhancer and SV40 poly A sequences. The light chain variable region for expression is inserted as a HindIII to BamHI fragment.

[0293] The DNA sequence is then to be confirmed to be correct for the humanized VH and VK in the expression vectors.

[0294] For antibody production the humanized heavy and light chain expression vectors may be introduced into appropriate production cell lines know in the art such as, for example, NS0 cells. Introduction of the expression vectors may be accomplished by co-transfection via electroporation or any other suitable transformation technology available in the art. Antibody producing cell lines can then be selected and expanded and humanized antibodies purified. The purified antibodies can then be analyzed by standard techniques such as SDS-PAGE.

ANTIBODY WITH IMPROVED AFFINITY, SPECIFICITY, STABILITY



[0295] The CDRL2 sequence ("KVSNRFS") of the mouse C2 antibody may be modified slightly without adversely affecting antibody activity. Conservative substitutions may be made through exchange of R for K at position 50 and S for N at position 53. The two alternative CDRL2 sequences are therefore "RVSNRFS" and "KVSSRFS", respectively. These are incorporated into the murine VK sequence with no other changes, as C2 VK-R and C2 VK-S, respectively.

[0296] The affinity, specificity and stability of an antibody according to the invention as described herein before or a fragment thereof can be modified by change of its glycosylation profile or pattern resulting in improved therapeutic values.

[0297] To achieve this change in glycosylation pattern, host cells may be engineered such that they are capable of expressing a preferred range of a glycoprotein-modifying glycosyl transferase activity which increases complex N-linked oligosaccharides carrying bisecting GIcNAc. Further, modified glycoforms of glycoproteins may be obtained, for example antibodies, including whole antibody molecules, antibody fragments, or fusion proteins that include a region equivalent to the Fc region of an immunoglobulin, having an enhanced Fc-mediated cellular cytotoxicity.

[0298] Methods of obtaining antibodies with modified glycosylation pattern are known to those skilled in the art and described, for example, in EP1071700, US2005272128, Ferrara et al (2006) J Biol Chem 281(8), 5032-5036); Ferrara et al (2006) Biotechnology and Bioengineering 93(5), 851-861.

PHARMACEUTICAL PREPARATION AND ADMINISTRATION



[0299] The antibodies according to the invention, but particularly a monoclonal antibody according the invention, can be prepared in a physiologically acceptable formulation and may comprise a pharmaceutically acceptable carrier, diluent and/or excipient using known techniques. For example, the antibody according to the invention and as described herein before in particular, the monoclonal antibody including any functional parts thereof is combined with a pharmaceutically acceptable carrier, diluent and/or excipient to form a therapeutic composition. Suitable pharmaceutical carriers, diluents and/or excipients are well known in the art and include, for example, phosphate buffered saline solutions, water, emulsions such as oil/water emulsions, various types of wetting agents, sterile solutions, etc.

[0300] Formulation of the pharmaceutical composition according to the invention can be accomplished according to standard methodology know to those skilled in the art.

[0301] The compositions of the present invention may be administered to a subject in the form of a solid, liquid or aerosol at a suitable, pharmaceutically effective dose. Examples of solid compositions include pills, creams, and implantable dosage units. Pills may be administered orally. Therapeutic creams may be administered topically. Implantable dosage units may be administered locally, for example, at a tumor site, or may be implanted for systematic release of the therapeutic composition, for example, subcutaneously. Examples of liquid compositions include formulations adapted for injection intramuscularly, subcutaneously, intravenously, intra-arterially, and formulations for topical and intraocular administration. Examples of aerosol formulations include inhaler formulations for administration to the lungs.

[0302] The compositions may be administered by standard routes of administration. In general, the composition may be administered by topical, oral, rectal, nasal, interdermal, intraperitoneal, or parenteral (for example, intravenous, subcutaneous, or intramuscular) routes. In addition, the composition may be incorporated into sustained release matrices such as biodegradable polymers, the polymers being implanted in the vicinity of where delivery is desired, for example, at the site of a tumor. The method includes administration of a single dose, administration of repeated doses at predetermined time intervals, and sustained administration for a predetermined period of time.

[0303] A sustained release matrix, as used herein, is a matrix made of materials, usually polymers which are degradable by enzymatic or acid/base hydrolysis or by dissolution. Once inserted into the body, the matrix is acted upon by enzymes and body fluids. The sustained release matrix desirably is chosen by biocompatible materials such as liposomes, polylactides (polylactide acid), polyglycolide (polymer of glycolic acid), polylactide co-glycolide (copolymers of lactic acid and glycolic acid), polyanhydrides, poly(ortho)esters, polypeptides, hyaluronic acid, collagen, chondroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, polyamino acids, amino acids such phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone. A preferred biodegradable matrix is a matrix of one of either polylactide, polyglycolide, or polylactide co-glycolide (co-polymers of lactic acid and glycolic acid).

[0304] It is well know to those skilled in the pertinent art that the dosage of the composition will depend on various factors such as, for example, the condition of being treated, the particular composition used, and other clinical factors such as weight, size, sex and general health condition of the patient, body surface area, the particular compound or composition to be administered, other drugs being administered concurrently, and the route of administration.

[0305] The composition may be administered in combination with other compositions comprising an biologically active substance or compound, particularly at least one compound selected from the group consisting of compounds against oxidative stress, anti-apoptotic compounds, metal chelators, inhibitors of DNA repair such as pirenzepin and metabolites, 3-amino-1-propanesulfonic acid (3APS), 1,3-propanedisulfonate (1,3PDS), α-secretase activators, β- and γ -secretase inhibitors, tau proteins, neurotransmitter, β-sheet breakers, attractants for amyloid beta clearing / depleting cellular components, inhibitors of N-terminal truncated amyloid beta including pyroglutamated amyloid beta 3-42, anti-inflammatory molecules, "atypical antipsychotics" such as, for example clozapine, ziprasidone, risperidone, aripiprazole or olanzapine or cholinesterase inhibitors (ChEIs) such as tacrine, rivastigmine, donepezil, and/or galantamine, M1 agonists and other drugs including any amyloid or tau modifying drug and nutritive supplements such as, for example, vitamin B12, cysteine, a precursor of acetylcholine, lecithin, choline, Ginkgo biloba, acyetyl-L-carnitine, idebenone, propentofylline, or a xanthine derivative, together with an antibody according to the present invention and, optionally, a pharmaceutically acceptable carrier and/or a diluent and/or an excipient and procedures for the treatment of diseases.

[0306] Proteinaceous pharmaceutically active matter may be present in amounts between 1 ng and 10 mg per dose. Generally, the regime of administration should be in the range of between 0.1 µg and 10 mg of the antibody according to the invention, particularly in a range 1.0 µg to 1.0 mg, and more particularly in a range of between 1.0 µg and 100 µg, with all individual numbers falling within these ranges also being part of the invention. If the administration occurs through continuous infusion a more proper dosage may be in the range of between 0.01 µg and 10 mg units per kilogram of body weight per hour with all individual numbers falling within these ranges also being part of the invention.

[0307] Administration will generally be parenterally, eg intravenously. Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions and emulsions. Non-aqueous solvents include without being limited to it, propylene glycol, polyethylene glycol, vegetable oil such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous solvents may be chosen from the group consisting of water, alcohol/aqueous solutions, emulsions or suspensions including saline and buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose) and others. Preservatives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, inert gases, etc.

[0308] The pharmaceutical composition may further comprise proteinaceous carriers such as, for example, serum albumin or immunoglobulin, particularly of human origin. Further biologically active agents may be present in the pharmaceutical composition of the invention dependent on its the intended use.

[0309] When the binding target is located in the brain, certain embodiments of the invention provide for the antibody or active fragment thereof to traverse the blood-brain barrier. Certain neurodegenerative diseases are associated with an increase in permeability of the blood-brain barrier, such that the antibody or active fragment thereof can be readily introduced to the brain. When the blood-brain barrier remains intact, several art-known approaches exist for transporting molecules across it, including, but not limited to, physical methods, lipid-based methods, and receptor and channel-based methods.

[0310] Physical methods of transporting the antibody or active fragment thereof across the blood-brain barrier include, but are not limited to, circumventing the blood-brain barrier entirely, or by creating openings in the blood-brain barrier. Circumvention methods include, but are not limited to, direct injection into the brain (see, e.g., Papanastassiou et al., Gene Therapy 9: 398-406 (2002)) and implanting a delivery device in the brain (see, e.g., Gill et al., Nature Med. 9: 589-595 (2003); and Gliadel Wafers™, Guildford Pharmaceutical). Methods of creating openings in the barrier include, but are not limited to, ultrasound (see, e.g., U.S. Patent Publication No. 2002/0038086), osmotic pressure (e.g., by administration of hypertonic mannitol (Neuwelt, E. A., Implication of the Blood-Brain Barrier and its Manipulation, Vols 1 & 2, Plenum Press, N.Y. (1989))), permeabilization by, e.g., bradykinin or permeabilizer A-7 (see, e.g., U.S. Patent Nos. 5,112,596, 5,268,164, 5,506,206, and 5,686,416), and transfection of neurons that straddle the blood-brain barrier with vectors containing genes encoding the antibody or antigen-binding fragment (see, e.g., U.S. Patent Publication No. 2003/0083299).

[0311] Lipid-based methods of transporting the antibody or active fragment thereof across the blood-brain barrier include, but are not limited to, encapsulating the antibody or active fragment thereof in liposomes that are coupled to antibody binding fragments that bind to receptors on the vascular endothelium of the blood-brain barrier (see, e.g., U.S. Patent Application Publication No. 20020025313), and coating the antibody or active fragment thereof in low-density lipoprotein particles (see, e.g., U.S. Patent Application Publication No. 20040204354) or apolipoprotein E (see, e.g., U.S. Patent Application Publication No. 20040131692).

[0312] Receptor and channel-based methods of transporting the antibody or active fragment thereof across the blood-brain barrier include, but are not limited to, using glucocorticoid blockers to increase permeability of the blood-brain barrier (see, e.g., U.S. Patent Application Publication Nos. 2002/0065259, 2003/0162695, and 2005/0124533); activating potassium channels (see, e.g., U.S. Patent Application Publication No. 2005/0089473), inhibiting ABC drug transporters (see, e.g., U.S. Patent Application Publication No. 2003/0073713); coating antibodies with a transferrin and modulating activity of the one or more transferrin receptors (see, e.g., U.S. Patent Application Publication No. 2003/0129186), and cationizing the antibodies (see, e.g., U.S. Patent No. 5,004,697).

DETECTION/DIAGNOSIS



[0313] In a further embodiment the present invention provides methods and kits for the detection and diagnosis of amyloid-associated diseases or conditions. These methods include known immunological methods commonly used for detecting or quantifying substances in biological samples or in an in situ condition.

[0314] Diagnosis of an amyloid-associated disease or condition in a patient may be achieved by detecting the immunospecific binding of a monoclonal antibody or an active fragment thereof to an epitope of the amyloid protein in a sample or in situ, which includes bringing the sample or a specific body part or body area suspected to contain the amyloid protein into contact with an antibody which binds an epitope of the amyloid protein, allowing the antibody to bind to the amyloid protein to form an immunological complex, detecting the formation of the immunological complex and correlating the presence or absence of the immunological complex with the presence or absence of amyloid protein in the sample or specific body part or area.

[0315] Biological samples that may be used in the diagnosis of an amyloid-associated disease or condition are, for example, fluids such as serum, plasma, saliva, gastric secretions, mucus, cerebrospinal fluid, lymphatic fluid and the like or tissue or cell samples obtained from an organism such as neural, brain, cardiac or vascular tissue. For determining the presence or absence of the amyloid protein in a sample any immunoassay known to those of ordinary skill in the art. (See Harlow and Lane, Antibodies: A Laboratory Manual (Cold Spring Harbor Laboratory, New York 1988 555-612) may be used such as, for example, assays which utilize indirect detection methods using secondary reagents for detection, ELISA's and immunoprecipitation and agglutination assays. A detailed description of these assays is, for example, given in WO96/13590 to Maertens and Stuyver, Zrein et al. (1998) and WO96/29605.

[0316] For in situ diagnosis, the antibody or any active and functional part thereof may be administered to the organism to be diagnosed by methods known in the art such as, for example, intravenous, intranasal, intraperitoneal, intracerebral, intraarterial injection such that a specific binding between the antibody according to the invention with an eptitopic region on the amyloid protein may occur. The antibody/antigen complex may be detected through a label attached to the antibody or a functional fragment thereof.

[0317] The immunoassays used in diagnostic applications typically rely on labelled antigens, antibodies, or secondary reagents for detection. These proteins or reagents can be labelled with compounds generally known to those skilled in the art including enzymes, radioisotopes, and fluorescent, luminescent and chromogenic substances including colored particles, such as colloidal gold and latex beads. Of these, radioactive labelling can be used for almost all types of assays and with most variations. Enzyme-conjugated labels are particularly useful when radioactivity must be avoided or when quick results are needed. Fluorochromes, although requiring expensive equipment for their use, provide a very sensitive method of detection. Antibodies useful in these assays include monoclonal antibodies, polyclonal antibodies, and affinity purified polyclonal antibodies.
Alternatively, the antibody may be labelled indirectly by reaction with labelled substances that have an affinity for immunoglobulin, such as protein A or G or second antibodies. The antibody may be conjugated with a second substance and detected with a labelled third substance having an affinity for the second substance conjugated to the antibody. For example, the antibody may be conjugated to biotin and the antibody-biotin conjugate detected using labelled avidin or streptavidin. Similarly, the antibody may be conjugated to a hapten and the antibody-hapten conjugate detected using labelled anti-hapten antibody.

[0318] Those of ordinary skill in the art will know of these and other suitable labels which may be employed in accordance with the present invention. The binding of these labels to antibodies or fragments thereof can be accomplished using standard techniques commonly known to those of ordinary skill in the art. Typical techniques are described by Kennedy, J. H., et al.,1976 (Clin. Chim. Acta 70:1-31), and Schurs, A. H. W. M., et al. 1977 (Clin. Chim Acta 81:1-40). Coupling techniques mentioned in the latter are the glutaraldehyde method, the periodate method, the dimaleimide method, and others.

[0319] Current immunoassays utilize a double antibody method for detecting the presence of an analyte, wherein. The antibody is labeled indirectly by reactivity with a second antibody that has been labeled with a detectable label. The second antibody is preferably one that binds to antibodies of the animal from which the monoclonal antibody is derived. In other words, if the monoclonal antibody is a mouse antibody, then the labeled, second antibody is an anti-mouse antibody. For the monoclonal antibody to be used in the assay described below, this label is preferably an antibody-coated bead, particularly a magnetic bead. For the polyclonal antibody to be employed in the immunoassay described herein, the label is preferably a detectable molecule such as a radioactive, fluorescent or an electrochemiluminescent substance.

[0320] An alternative double antibody system often referred to as fast format systems because they are adapted to rapid determinations of the presence of an analyte, may also be employed within the scope of the present invention. The system requires high affinity between the antibody and the analyte. According to one embodiment of the present invention, the presence of the amyloid protein is determined using a pair of antibodies, each specific for amyloid protein. One of said pairs of antibodies is referred to herein as a "detector antibody" and the other of said pair of antibodies is referred to herein as a "capture antibody". The monoclonal antibody of the present invention can be used as either a capture antibody or a detector antibody. The monoclonal antibody of the present invention can also be used as both capture and detector antibody, together in a single assay. One embodiment of the present invention thus uses the double antibody sandwich method for detecting amyloid protein in a sample of biological fluid. In this method, the analyte (amyloid protein) is sandwiched between the detector antibody and the capture antibody, the capture antibody being irreversibly immobilized onto a solid support. The detector antibody would contain a detectable label, in order to identify the presence of the antibody-analyte sandwich and thus the presence of the analyte.

[0321] Exemplary solid phase substances include, but are not limited to, microtiter plates, test tubes of polystyrene, magnetic, plastic or glass beads and slides which are well known in the field of radioimmunoassay and enzyme immunoassay. Methods for coupling antibodies to solid phases are also well known to those skilled in the art. More recently, a number of porous material such as nylon, nitrocellulose, cellulose acetate, glass fibers and other porous polymers have been employed as solid supports.

[0322] The present invention also relates to a diagnostic kit for detecting amyloid protein in a biological sample comprising a composition as defined above. Moreover, the present invention relates to the latter diagnostic kit which, in addition to a composition as defined above, also comprises a detection reagent as defined above. The term "diagnostic kit" refers in general to any diagnostic kit known in the art. More specifically, the latter term refers to a diagnostic kit as described in Zrein et al. (1998).

[0323] It is still another object of the present invention to provide novel immunoprobes and test kits for detection and diagnosis of amyloid-associated diseases and conditions comprising antibodies according to the present invention. For immunoprobes, the antibodies are directly or indirectly attached to a suitable reporter molecule, e.g., an enzyme or a radionuclide. The test kit includes a container holding one or more antibodies according to the present invention and instructions for using the antibodies for the purpose of binding to amyloid protein to form an immunological complex and detecting the formation of the immunological complex such that presence or absence of the immunological complex correlates with presence or absence of amyloid protein.

EXAMPLES


Materials



[0324] The development and preparation of mouse monoclonal antibody ACI-01-Ab7C2 (named "mC2" and hC2 for the humanized C2 antibody, throughout the application) is described in co-pending application EP 05 02 7092.5 filed 12.12.2005.

[0325] Hybridoma cells FP-12H3-C2, producing mouse monoclonal antibody ACI-01-Ab7C2 (named "mC2" and hC2 for the humanized C2 antibody, throughout the application) were deposited 01 December 2005 in co-pending application no EP05027092.5 with the "Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) in Braunschweig, Mascheroder Weg 1 B, 38124 Braunschweig, under the provisions of the Budapest Treaty and given accession no DSM ACC2750.

[0326] Hybridoma cells were cultured in Dulbecco's modified Eagle Medium (DMEM) supplemented with 10% foetal bovine serum and antibiotics (Penicillin/Streptomycin). The isotype of the antibody produced was checked and found to be mouse IgG2b/kappa, as expected.

Assay



[0327] An ELISA for binding to Amyloid Beta provided a reliable measure of the potency of C2 antibodies. Positive control antibodies, murine FP-12H3-C2 antibody (Genovac Lot No: AK379/01), and standard Chemicon antibody 1560 (Lot no: 0508008791).

Choice of human constant regions



[0328] As immune system recruitment is not desirable for the clinical antibody candidate, the selected human constant region for the heavy chain was human IgG4, modified to change Serine at position 228 in the hinge region to Proline (HuIgG4 Ser-Pro). This mutation stabilizes the interchain disulphide bond and prevents the formation of half molecules that may occur in native human IgG4 preparations. The antibody expressed from the production cell lines will also have the terminal lysine removed. The sequences of human constant regions HuIgG4 Ser-Pro and human Kappa are given in SEQ ID NO: 17 and 14, respectively.

Sequence Listing



[0329] 

<110> AC Immune S.A.

<110> Genentech, Inc.

<120> Humanized Antibody

<130> M1967 EP/A/1 BS

<140> EP 07840408.4
<141> 2007-07-13

<150> EP 06014730.3
<151> 2006-07-14

<150> EP 06020765.1
<151> 2006-10-02

<150> US 60/943,289
<151> 2007-06-11

<150> US 60/943,499
<151> 2007-06-12

<160> 32

<170> PatentIn version 3.3

<210> 1
<211> 10
<212> PRT
<213> Mus musculus

<220>
<223> C2 HuVH AF 4 humanised heavy chain variable region (CDR1)

<400> 1

<210> 2
<211> 17
<212> PRT
<213> Mus musculus

<220>
<223> C2 HuVH AF 4 humanised heavy chain variable region (CDR2)

<400> 2

<210> 3
<211> 3
<212> PRT
<213> Mus musculus

<220>
<223> C2 HuVH AF 4 humanised heavy chain variable region (CDR3)

<400> 3

<210> 4
<211> 16
<212> PRT
<213> Mus musculus

<220>
<223> C2 HuVK 1 humanised light chain variable region (CDR1)

<400> 4

<210> 5
<211> 7
<212> PRT
<213> Mus musculus

<220>
<223> C2 HuVK 1 humanised light chain variable region (CDR2)

<400> 5

<210> 6
<211> 9
<212> PRT
<213> Mus musculus

<220>
<223> C2 HuVK 1 humanised light chain variable region (CDR3)

<400> 6

<210> 7
<211> 6
<212> PRT
<213> Homo sapiens

<220>
<223> A-betta epitope, region 2

<400> 7

<210> 8
<211> 5
<212> PRT
<213> Homo sapiens

<220>
<223> A-beta epitope, region 1

<400> 8

<210> 9
<211> 6
<212> PRT
<213> Homo sapiens

<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be Ala, Val, Leu, norleucine, Met, Phe, or Ile

<220>
<221> misc_feature
<222> (4)..(4)
<223> Xaa can be Ala, Val, Leu, Ser, or Ile

<220>
<221> misc_feature
<222> (5)..(5)
<223> Xaa can be Glu or Asp

<220>
<221> misc_feature
<222> (6)..(6)
<223> Xaa can be Glu or Asp

<400> 9

<210> 10
<211> 5
<212> PRT
<213> Homo sapiens

<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be His, Asn, Gln, Lys, or Arg

<220>
<221> misc_feature
<222> (2)..(2)
<223> Xaa can be Asn or Gln

<220>
<221> misc_feature
<222> (5)..(5)
<223> Xaa can be Ala, Val, Leu, norleucine, Met, Phe, or Ile

<400> 10

<210> 11
<211> 10
<212> PRT
<213> Homo sapiens

<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be His, Asn, Gln, Lys, or Arg

<220>
<221> misc_feature
<222> (2)..(2)
<223> Xaa can be Asn or Gln

<220>
<221> misc_feature
<222> (5)..(5)
<223> Xaa can be Ala, Val, Leu, norleucine, Met, Phe, or Ile

<220>
<221> misc_feature
<222> (8)..(8)
<223> Xaa can be Ala, Val, Leu, Ser, or Ile

<220>
<221> misc_feature
<222> (9)..(9)
<223> Xaa can be Glu or Asp

<220>
<221> misc_feature
<222> (10)..(10)
<223> Xaa can be Glu or Asp

<400> 11

<210> 12
<211> 112
<212> PRT
<213> Artificial sequence

<220>
<223> artificial humanized C2 HuVK 1 variable light chain

<400> 12

<210> 13
<211> 219
<212> PRT
<213> artificial sequence

<220>
<223> artificial humanized C2 light chain

<400> 13



<210> 14
<211> 107
<212> PRT
<213> artificial sequence

<220>
<223> artificial humanized C2 light chain constant region

<400> 14

<210> 15
<211> 112
<212> PRT
<213> artificial sequence

<220>
<223> artificial humanized C2 HuVH AF 4 variable heavy chain

<400> 15

<210> 16
<211> 439
<212> PRT
<213> artificial sequence

<220>
<223> artificial humanized C2 heavy chain

<400> 16





<210> 17
<211> 326
<212> PRT
<213> Homo sapiens

<220>
<223> IG GAMMA-4 CHAIN C REGION - modified

<400> 17



<210> 18
<211> 51
<212> DNA
<213> Mus musculus

<220>
<223> C2 HuVH AF 4 humanised heavy chain variable region (CDR2)

<400> 18
agcatcaata gtaatggtgg tagcacctat tatccagaca gtgtgaaggg c   51

<210> 19
<211> 9
<212> DNA
<213> Mus musculus

<220>
<223> C2 HuVH AF 4 humanised heavy chain variable region (CDR3)

<400> 19
ggtgactac   9

<210> 20
<211> 49
<212> DNA
<213> Mus musculus

<220>
<223> C2 HuVK 1 humanised light chain variable region (CDR1)

<400> 20
agatctagtc agagccttgt atatagtaat ggagacacct atttacatt   49

<210> 21
<211> 336
<212> DNA
<213> artificial sequence

<220>
<223> artificial humanized C2 Hu VK 1 variable light chain

<400> 21

<210> 22
<211> 657
<212> DNA
<213> artificial sequence

<220>
<223> artificial humanized C2 light chain

<400> 22

<210> 23
<211> 321
<212> DNA
<213> Homo sapiens

<220>
<223> artificial humanized C2 light chain constant region

<400> 23

<210> 24
<211> 336
<212> DNA
<213> artificial sequence

<220>
<223> artificial humanized C2 HuVH AF variable heavy chain

<400> 24

<210> 25
<211> 1317
<212> DNA
<213> artificial sequence

<220>
<223> artificial humanized C2 heavy chain

<400> 25

<210> 26
<211> 981
<212> DNA
<213> Homo sapiens

<220>
<223> artificial humanized C2 heavy chain constant region

<400> 26

<210> 27
<211> 112
<212> PRT
<213> Mus musculus

<400> 27



<210> 28
<211> 112
<212> PRT
<213> Mus musculus

<400> 28

<210> 29
<211> 336
<212> DNA
<213> Mus musculus

<400> 29

<210> 30
<211> 417
<212> DNA
<213> Mus musculus

<400> 30

<210> 31
<211> 336
<212> DNA
<213> Mus musculus

<400> 31

<210> 32
<211> 408
<212> DNA
<213> Mus musculus

<400> 32




Claims

1. A humanized antibody or a fragment thereof, wherein said antibody or fragment thereof comprises:

(A) (i) a Heavy Chain Variable Region (HCVR) having an amino acid sequence that is 96%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 15, wherein the HCVR comprises a heavy chain complementarity determining region 1 (CDR1) as given in SEQ ID NO: 1, a heavy chain CDR2 as given in SEQ ID NO: 2 and a heavy chain CDR3 as given in SEQ ID NO: 3, and (ii) a light chain variable region (LCVR) comprising a light chain CDR1 as given in SEQ ID NO: 4, a light chain CDR2 as given in SEQ ID NO: 5, RVSNRFS or KVSSRFS and a light chain CDR3 as given in SEQ ID NO: 6, or

(B) (i) an HCVR comprising a heavy chain CDR1 as given in SEQ ID NO: 1, a heavy chain CDR2 as given in SEQ ID NO: 2 and a heavy chain CDR3 as given in SEQ ID NO: 3, and (ii) a LCVR having an amino acid sequence that is 96%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 12, wherein the LCVR comprises a light chain CDR1 as given in SEQ ID NO: 4, a light chain CDR2 as given in SEQ ID NO: 5, RVSNRFS or KVSSRFS and a light chain CDR3 as given in SEQ ID NO: 6,

wherein said antibody or fragment thereof specifically binds to β-amyloid protein.
 
2. The humanized antibody or fragment thereof according to claim 1(A), wherein the LCVR has an amino acid sequence that is 96%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 12.
 
3. The humanized antibody or fragment thereof according to claim 1(B), wherein the HCVR has an amino acid sequence that is 96%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 15.
 
4. The humanized antibody or a fragment thereof according to any one of claims 1-3, which comprises:

i) the HCVR having the amino acid sequence of SEQ ID NO: 15 or an amino acid sequence that is 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 15; or

ii) the LCVR having the amino acid sequence of SEQ ID NO: 12 or an amino acid sequence that is 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 12; or

iii) the HCVR having the amino acid sequence of SEQ ID NO: 15 or an amino acid sequence that is 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 15 and the LCVR having the amino acid sequence of SEQ ID NO: 12 or an amino acid sequence that is 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 12.


 
5. The humanized antibody or fragment thereof according to any one of claims 1-3, which comprises:

i) a heavy chain comprising the amino acid sequence of SEQ ID NO: 16 or an amino acid sequence that is 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 16; or

ii) a light chain comprising the amino acid sequence of SEQ ID NO: 13 or an amino acid sequence that is 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 13; or

iii) a heavy chain comprising the amino acid sequence of SEQ ID NO: 16 or an amino acid sequence that is 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 16 and a light chain comprising the amino acid sequence of SEQ ID NO: 13 or an amino acid sequence that is 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 13.


 
6. The humanized antibody or fragment thereof according to any one of the preceding claims, which antibody is of the IgG4 isotype.
 
7. The humanized antibody or fragment thereof according to any one of the preceding claims, wherein said humanized antibody or fragment thereof is capable of inhibiting the aggregation of β-amyloid monomers to high molecular polymeric fibrils and of disaggregating preformed polymeric fibrils or filaments.
 
8. The humanized antibody or fragment thereof according to any one of the preceding claims, wherein said antibody or fragment thereof binds to an Aβ fiber, fibril or filament with a binding affinity that is at least 10 times, at least 15 times, at least 20 times, or at least 25 times, higher than the binding affinity of said antibody or fragment thereof to an Aβ monomer.
 
9. The humanized antibody or fragment thereof according to any one of the preceding claims, wherein

(a) the amino acid in Kabat position 47 of the framework regions of the Heavy Chain Variable Region is the amino acid Leu; and/or

(b) the amino acid in Kabat position 94 in the framework regions of the Heavy Chain Variable Region is the amino acid Ser; and/or

(c) the amino acid in Kabat position 87 in the framework regions of the Light Chain Variable Region is the amino acid Tyr, Phe, Leu, VaI, Ile, or Ala.


 
10. A nucleic acid molecule comprising a nucleotide sequence encoding the humanized antibody or fragment thereof according to any one of claims 1-9.
 
11. An expression vector comprising the nucleic acid molecule of claim 10.
 
12. A cell comprising the expression vector of claim 11.
 
13. A composition comprising the humanized antibody or fragment thereof according to any one of claims 1-9, and further comprising a pharmaceutically acceptable carrier, a diluent, and/or an excipient.
 
14. A mixture comprising the humanized antibody or fragment thereof according to any one of claims 1-9, and further comprising a biologically active substance, a pharmaceutically acceptable carrier, a diluent, and/or an excipient.
 
15. The mixture according to claim 14, comprising:

(i) a biologically active substance that is a compound used in the treatment of amyloidosis, or

(ii) at least one of the following compounds: an anti-oxidative stress compound; an anti-apoptotic compound; a metal chelator; a DNA repair inhibitor such as pirenzepine and metabolites; 3-amino-1-propanesulfonic acid (3 APS); 1,3-propanedisulfonate (1,3PDS); α-secretase activator; a β-secretase inhibitor; a γ-secretase inhibitor; a tau protein; a neurotransmitter; a β-sheet breaker; an attractant for amyloid beta clearing / depleting cellular components; an inhibitor of N-terminal truncated amyloid beta, such as pyroglutamated amyloid beta 3-42; an anti-inflammatory molecule; a cholinesterase inhibitor (ChEI) such as tacrine, rivastigmine, donepezil, and/or galantamine; an Ml agonist; or another drug such as an amyloid- or tau-modifying drug or nutritive supplement.


 
16. A humanized antibody or fragment thereof as defined in any one of claims 1-9, a composition as defined in claim 13, or a mixture as defined in claim 14 or 15 for use in a method of treating or alleviating the effects of amyloidosis in an animal, such as a mammal or a human.
 
17. The humanized antibody or fragment thereof, the mixture, or the composition for use according to claim 16, wherein the amyloidosis is secondary amyloidosis, age-related amyloidosis, a neurological disorder, Alzheimer's Disease (AD), Lewy body dementia, Down's syndrome, hereditary cerebral hemorrhage with amyloidosis (Dutch type), the Guam Parkinson-Dementia complex, progressive supranuclear palsy, multiple sclerosis, Creutzfeld Jacob disease, Parkinson's disease, HIV-related dementia, ALS (amyotropic lateral sclerosis), Adult Onset Diabetes, senile cardiac amyloidosis, endocrine tumors, or macular degeneration.
 
18. The humanized antibody or fragment thereof, the mixture, or the composition for use according to claim 17, wherein the amyloidosis is Alzheimer's disease.
 
19. The humanized antibody or fragment thereof, the mixture, or the composition for use according to claim 16, wherein the treatment of the animal leads to

i) an increase of the cognitive memory capacity and/or;

ii) the retention of cognitive memory capacity; and/or

iii) a complete restoration of the cognitive memory capacity.


 
20. A method of diagnosis of an amyloidosis in a patient comprising:

i) contacting a sample, body part, or body area containing an amyloid protein with the humanized antibody or fragment thereof according to any one of claims 1-9 under conditions that allow binding of said antibody or fragment thereof to the amyloid protein; and

ii) detecting the antibody or fragment thereof bound to the protein,

wherein the presence or absence of antibody or fragment thereof bound to the amyloid protein indicates the presence or absence of amyloid protein in said sample, body part, or body area.
 
21. A method of determining the extent of amyloidogenic plaque burden in a tissue sample or body fluid sample comprising:

i) testing a tissue sample or body fluid sample for the presence of amyloid protein with the humanized antibody or fragment thereof according to any one of claims 1-9;

ii) determining the amount of antibody or fragment thereof bound to the amyloid protein; and

iii) calculating the plaque burden in the tissue sample or body fluid sample.


 
22. A kit for detection and diagnosis of amyloid-associated diseases and conditions comprising the humanized antibody or fragment thereof according to any one of claims 1-9, wherein said kit comprises a container holding one or more antibodies or fragments thereof according to any one of claims 1-9 and instructions for using said antibodies or fragments thereof.
 
23. A humanized antibody or fragment thereof as defined in any one of claims 1-9, a composition as defined in claim 13, or a mixture as defined in claim 14 or 15, for use in a method of preventing degeneration of neurons upon exposure to β-amyloid oligomers, or for use in a method of protecting neurons from β-amyloid-induced degradation.
 
24. A method for preparing a humanized antibody or fragment thereof as defined in any one of claims 1-9, wherein the method comprises culturing the cell of claim 12, and purifying the antibody or fragment thereof.
 


Ansprüche

1. Humanisierter Antikörper oder ein Fragment davon, wobei der Antikörper oder das Fragment davon umfasst:

(A) (i) eine variable Domäne der schweren Kette (HCVR) mit einer Aminosäuresequenz, die mit der Aminosäuresequenz von SEQ ID NO: 15 zu 96%, 97%, 98%, 99% oder 100% identisch ist, wobei die HCVR eine schwere Kette-Komplementarität-Bestimmungs-Domäne 1 (CDR1), wie in SEQ ID NO: 1 angegeben, eine schwere Kette CDR2, wie in SEQ ID NO: 2 angegeben und ein schwere Kette CDR3, wie in SEQ ID NO: 3 angegeben, umfasst und (ii) eine variable Domäne der leichten Kette (LCVR), umfassend eine leichte Kette CDR1, wie in SEQ ID NO: 4 angegeben, eine leichte Kette CDR2, wie in SEQ ID NO: 5 angegeben, RVSNRFS oder KVSSRFS und eine leichte Kette CDR3, wie in SEQ ID NO: 6 angegeben, oder

(B) (i) eine HCVR, umfassend eine schwere Kette CDR1, wie in SEQ ID NO: 1 angegeben, eine schwere Kette CDR2, wie in SEQ ID NO: 2 angegeben und ein schwere Kette CDR3, wie in SEQ ID NO: 3 angegeben, und (ii) eine LCVR mit einer Aminosäuresequenz, die mit der Aminosäuresequenz von SEQ ID NO: 12 zu 96%, 97%, 98%, 99% oder 100% identisch ist, wobei die LCVR eine leichte Kette CDR1, wie in SEQ ID NO: 4 angegeben, eine leichte Kette CDR2, wie in SEQ ID NO: 5 angegeben, RVSNRFS oder KVSSRFS und eine leichte Kette CDR3, wie in SEQ ID NO: 6 angegeben, umfasst,

wobei der Antikörper oder das Fragment davon spezifisch an β-Amyloidprotein bindet.
 
2. Humanisierter Antikörper oder das Fragment davon nach Anspruch 1(A), wobei die LCVR eine Aminosäuresequenz, die mit der Aminosäuresequenz von SEQ ID NO: 12 zu 96%, 97%, 98%, 99% oder 100% identisch ist, aufweist.
 
3. Humanisierter Antikörper oder das Fragment davon nach Anspruch 1(B), wobei die HCVR eine Aminosäuresequenz, die mit der Aminosäuresequenz von SEQ ID NO: 15 zu 96%, 97%, 98%, 99% oder 100% identisch ist, aufweist.
 
4. Humanisierter Antikörper oder ein Fragment davon nach einem der Ansprüche 1-3, der umfasst:

i) die HCVR mit der Aminosäuresequenz von SEQ ID NO: 15 oder einer Aminosäuresequenz, die mit der Aminosäuresequenz von SEQ ID NO: 15 zu 97%, 98%, oder 99% identisch ist; oder

ii) die LCVR mit der Aminosäuresequenz von SEQ ID NO: 12 oder einer Aminosäuresequenz, die mit der Aminosäuresequenz von SEQ ID NO: 12 zu 97%, 98%, oder 99% identisch ist; oder

iii) die HCVR mit der Aminosäuresequenz von SEQ ID NO: 15 oder einer Aminosäuresequenz, die mit der Aminosäuresequenz von SEQ ID NO: 15 zu 97%, 98%, oder 99% identisch ist und die LCVR mit der Aminosäuresequenz von SEQ ID NO: 12 oder einer Aminosäuresequenz, die mit der Aminosäuresequenz von SEQ ID NO: 12 zu 97%, 98%, oder 99% identisch ist.


 
5. Humanisierter Antikörper oder das Fragment davon nach einem der Ansprüche 1-3, der umfasst:

i) eine schwere Kette, umfassend die Aminosäuresequenz von SEQ ID NO: 16 oder eine Aminosäuresequenz, die mit der Aminosäuresequenz von SEQ ID NO: 16 zu 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, oder 99% identisch ist; oder

ii) eine leichte Kette, die die Aminosäuresequenz von SEQ ID NO: 13 oder eine Aminosäuresequenz, die mit der Aminosäuresequenz von SEQ ID NO: 13 zu 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, oder 99% identisch ist, umfasst; oder

iii) eine schwere Kette, umfassend die Aminosäuresequenz von SEQ ID NO: 16 oder eine Aminosäuresequenz, die mit der Aminosäuresequenz von SEQ ID NO: 16 zu 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, oder 99% identisch ist und eine leichte Kette, umfassend die Aminosäuresequenz von SEQ ID NO: 13 oder eine Aminosäuresequenz, die mit der Aminosäuresequenz von SEQ ID NO: 13 zu 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, oder 99% identisch ist.


 
6. Humanisierter Antikörper oder das Fragment davon nach einem der vorstehenden Ansprüche, wobei der Antikörper vom Isotyp IgG4 ist.
 
7. Humanisierter Antikörper oder das Fragment davon nach einem der vorstehenden Ansprüche, wobei der humanisierte Antikörper oder das Fragment davon zur Hemmung der Aggregation von β-Amyloid-Monomeren an polymere Fibrillen mit hohem Molekulargewicht und Disaggregieren von vorgebildeten polymeren Fibrillen oder Filamenten befähigt ist.
 
8. Humanisierter Antikörper oder das Fragment davon nach einem der vorstehenden Ansprüche, wobei der Antikörper oder das Fragment davon an eine Aß-Faser, Fibrille oder Filament mit einer Bindungsaffinität bindet, die mindestens 10 mal, mindestens 15 mal, mindestens 20 mal oder mindestens 25 mal höher als die Bindungsaffinität von dem Antikörper oder Fragment davon an ein Aß-Monomer ist.
 
9. Humanisierter Antikörper oder das Fragment davon nach einem der vorstehenden Ansprüche, wobei

(a) die Aminosäure in Kabat-Position 47 von den Framework-Domänen der variablen Domäne der schweren Kette die Aminosäure Leu ist; und/oder

(b) die Aminosäure in Kabat-Position 94 in den Framework-Domänen der variablen Domäne der schweren Kette die Aminosäure Ser ist; und/oder

(c) die Aminosäure in Kabat-Position 87 in den Framework-Domänen der variablen Domäne der leichten Kette die Aminosäure Tyr, Phe, Leu, Val, Ile oder Ala ist.


 
10. Nukleinsäuremolekül, umfassend eine Nukleotidsequenz, kodierend für den humanisierten Antikörper oder das Fragment davon nach einem der Ansprüche 1-9.
 
11. Expressionsvektor, umfassend das Nukleinsäuremolekül nach Anspruch 10.
 
12. Zelle, umfassend den Expressionsvektor nach Anspruch 11.
 
13. Zusammensetzung, umfassend den humanisierten Antikörper oder das Fragment davon nach einem der Ansprüche 1-9, und weiterhin umfassend einen pharmazeutisch akzeptablen Träger, ein Verdünnungsmittel und/oder einen Hilfsstoff.
 
14. Gemisch, umfassend den humanisierten Antikörper oder das Fragment davon nach einem der Ansprüche 1-9, und weiterhin umfassend eine biologisch aktive Substanz, einen pharmazeutisch akzeptablen Träger, ein Verdünnungsmittel und/oder einen Hilfsstoff.
 
15. Gemisch nach Anspruch 14, umfassend:

(i) eine biologisch aktive Substanz, die eine Verbindung ist, welche zur Behandlung von Amyloidose verwendet wird, oder

(ii) mindestens eine der folgenden Verbindungen: eine Verbindung gegen oxidativen Stress; eine anti-apoptotische Verbindung; einen Metallchelator; einen Inhibitor der DNS-Reparatur, wie Pirenzepin und Metabolite; 3-Amino-1-propansulfonsäure (3 APS); 1,3-Propandisulfonat (1,3PDS); α-Sekretaseaktivator; β-Sekretaseinhibitor; einen γ-Sekretaseinhibitor; ein tau-Protein; einen Neurotransmitter; einen β-Faltblattunterbrecher; ein Lockmittel für Amyloid-beta zur Abtrennung/Entsorgung zellulärer Komponenten; einen Inhibitor von N-terminal trunkiertem Amyloid-beta, wie pyroglutamiertes Amyloid-beta 3-32; ein entzündungshemmendes Mittel; einen Cholinesteraseinhibitor (ChEI), wie Tacrin, Rivastigmin, Donepezil und/oder Galantamin; einen M1-Agonisten oder ein anderes Arzneimittel, wie ein Amyloid- oder tau-modifizierendes Arzneimittel oder Nahrungsergänzungsmittel.


 
16. Humanisierter Antikörper oder ein Fragment davon, wie in einem der Ansprüche 1-9 definiert, eine Zusammensetzung, wie in Anspruch 13 definiert, oder ein Gemisch, wie in Anspruch 14 oder 15 definiert, zur Verwendung in einem Verfahren zur Behandlung oder Linderung der Auswirkungen von Amyloidose in einem Tier, wie ein Säugetier oder Mensch.
 
17. Humanisierter Antikörper oder das Fragment davon, das Gemisch oder die Zusammensetzung zur Verwendung nach Anspruch 16, wobei die Amyloidose sekundäre Amyloidose, altersbedingte Amyloidose, eine neurologische Störung, Alzheimersche Krankheit (AD), Lewy-Körper-Demenz, Down-Syndrom, angeborene cerebrale Hämorrhagie mit Amyloidose (Dutch type); der Guam-Parkinson-Demenz-Komplex; progressive supranukleare Blickparese, multiple Sklerose; Creutzfeld-Jacob-Krankheit, Parkinson-Krankheit, HIV-induzierte Demenz, ALS (amyotrophe Lateralsklerose), Altersdiabetes; senile kardiale Amyloidose; endokrine Tumore und Makuladegeneration ist.
 
18. Humanisierter Antikörper oder das Fragment davon, das Gemisch oder die Zusammensetzung zur Verwendung nach Anspruch 17, wobei die Amyloidose Alzheimersche Krankheit ist.
 
19. Humanisierter Antikörper oder das Fragment davon, das Gemisch oder die Zusammensetzung zur Verwendung nach Anspruch 16, wobei die Behandlung des Tieres führt zu

i) einer Erhöhung des kognitiven Erinnerungsvermögens und/oder;

ii) der Beibehaltung des kognitiven Erinnerungsvermögens; und/oder

iii) einer kompletten Wiederherstellung des kognitiven Erinnerungsvermögens.


 
20. Verfahren zur Diagnose einer Amyloidose bei einem Patienten, umfassend:

i) Kontaktieren einer Probe, eines Körperteils oder einer Körperfläche, die ein Amyloidprotein enthält, mit dem humanisierten Antikörper oder dem Fragment davon nach einem der Ansprüche 1-9 unter Bedingungen, die das Binden des Antikörpers oder Fragments davon an das Amyloidprotein erlauben; und

ii) Nachweisen des Antikörpers oder Fragments davon, gebunden an das Protein, wobei die Anwesenheit oder Abwesenheit von Antikörper oder Fragment davon, gebunden an das Amyloidprotein, die Anwesenheit oder Abwesenheit von Amyloidprotein in der Probe, dem Körperteil oder der Körperfläche anzeigt.


 
21. Verfahren zur Bestimmung des Ausmaßes der Belastung mit amyloidogenen Plaque in einer Gewebeprobe oder Körperflüssigkeitsprobe, umfassend:

i) Testen einer Gewebeprobe oder Körperflüssigkeitsprobe auf die Anwesenheit von Amyloidprotein mit dem humanisierten Antikörper oder dem Fragment davon nach einem der Ansprüche 1-9;

ii) Bestimmung der Menge von Antikörper oder dem Fragment davon, gebunden an das Amyloidprotein; und

iii) Ausrechnen der Plaquebelastung in der Gewebeprobe oder Körperflüssigkeitsprobe.


 
22. Kit zum Nachweis und zur Diagnose von Amyloid-assoziierten Krankheiten und Zuständen, umfassend den humanisierten Antikörper oder das Fragment davon nach einem der Ansprüche 1-9, wobei das Kit einen Container, der einen oder mehrere Antikörper oder Fragmente davon nach einem der Ansprüche 1-9 beinhaltet, und Anweisungen zur Verwendung der Antikörper oder Fragmente davon umfasst.
 
23. Humanisierter Antikörper oder ein Fragment davon, wie in einem der Ansprüche 1-9 definiert, eine Zusammensetzung, wie in Anspruch 13 definiert, oder ein Gemisch, wie in Anspruch 14 oder 15 definiert, zur Verwendung in einem Verfahren zur Vorbeugung einer Neuronendegeneration während einer Exposition zu β-Amyloidoligomeren oder zur Verwendung in einem Verfahren zum Schützen von Neuronen vor β-Amyloid-induzierter Degradation.
 
24. Verfahren zum Herstellen eines humanisierten Antikörpers oder eines Fragments davon, wie in einem der Ansprüche 1-9 definiert, wobei das Verfahren Züchten der Zelle von Anspruch 12, und Reinigen des Antikörpers oder Fragments davon umfasst.
 


Revendications

1. Anticorps humanisé ou un fragment de celui-ci, dans lequel ledit anticorps ou un fragment de celui-ci comprend:

(A)

(i) une région variable de chaîne lourde (HCVR) ayant une séquence d'acides aminés qui est identique à 96%, 97%, 98%, 99% ou 100% à la séquence d'acides aminés de SEQ ID NO: 15; dans lequel HCRV comprend une région déterminant la complémentarité d'une chaîne lourde (CDR1) comme indiqué dans SEQ ID NO: 1, une chaîne lourde CDR2 comme indiqué dans SEQ ID NO: 2 et une chaîne lourde CDR3 comme indiqué dans SEQ ID NO:3, et

(ii) une région variable de chaîne légère (LCVR) comprenant une chaîne légère CDR1 comme indiqué dans SEQ ID NO:4, une chaîne légère CDR2 comme indiqué dans SEQ ID NO:5, RVSNRFS ou KVSSRFS et une chaîne légère CDR3 comme indiqué dans SEQ ID NO:6, ou

(B)

(i) HCRV comprenant une chaîne lourde CDR1 comme indiqué dans SEQ ID NO: 1, une chaîne lourde CDR2 comme indiqué dans SEQ ID NO: 2 et une chaîne lourde CDR3 comme indiqué dans SEQ ID NO: 3 et

(ii) LCVR ayant une séquence d'acides aminés qui est identique à 96%, 97%, 98%, 99% ou 100% à la séquence d'acides aminés SEQ ID NO: 12, dans lequel LCVR comprend une chaîne légère CDR1 comme indiqué dans SEQ ID NO:4, une chaîne légère CDR2 comme indiqué dans SEQ ID NO:5, RVSNRFS ou KVSSRFS et une chaîne légère CDR3 comme indiqué dans SEQ ID NO:6,

dans lequel ledit anticorps ou un fragment de celui-ci se lie spécifiquement à la protéine β-amyloïde.


 
2. Anticorps humanisé ou un fragment de celui-ci selon la revendication 1(A), dans lequel LCVR a une séquence d'acides aminés qui est identique à 96%, 97%, 98%, 99% ou 100% à la séquence d'acides aminés SEQ ID NO: 12.
 
3. Anticorps humanisé ou un fragment de celui-ci selon la revendication 1(B), dans lequel la HCVR a une séquence d'acides aminés qui est identique à 96%, 97%, 98%, 99% ou 100% à la séquence d'acides aminés SEQ ID NO: 15.
 
4. Anticorps humanisé ou un fragment de celui-ci selon l'une quelconque des revendications 1 à 3, qui comprend:

i) HCVR ayant la séquence d'acides aminés SEQ ID NO: 15 ou une séquence d'acides aminés qui est identique à 97%, 98%, ou 99% à la séquence d'acides aminés SEQ ID NO: 15 ; ou

ii) LCVR ayant la séquence d'acides aminés SEQ ID NO: 12 ou une séquence d'acides aminés qui est identique à 97%, 98%, ou 99% à la séquence d'acides aminés SEQ ID NO: 12 ; ou

iii) HCVR ayant la séquence d'acides aminés SEQ ID NO: 15 ou une séquence d'acides aminés qui est identique à 97%, 98%, ou 99% à la séquence d'acides aminés de SEQ ID NO: 15 et LCVR ayant la séquence d'acides aminés SEQ ID NO: 12 ou une séquence d'acides aminés qui est identique à 97%, 98%, ou 99% à la séquence d'acides aminés SEQ ID NO: 12.


 
5. Anticorps humanisé ou un fragment de celui-ci selon l'une quelconque des revendications 1 à 3, qui comprend:

i) une chaîne lourde comprenant la séquence d'acides aminés SEQ ID NO: 16 ou une séquence d'acides aminés qui est identique à 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, ou 99% à la séquence d'acides aminés SEQ ID NO: 16; ou

ii) une chaîne légère comprenant la séquence d'acides aminés SEQ ID NO: 13 ou une séquence d'acides aminés qui est identique à 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, ou 99% à la séquence d'acides aminés SEQ ID NO: 13; ou

iii) une chaîne lourde comprenant la séquence d'acides aminés SEQ ID NO: 16 ou une séquence d'acides aminés qui est identique à 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, ou 99% à la séquence d'acides aminés SEQ ID NO: 16 et une chaîne légère comprenant une séquence d'acides aminés SEQ ID NO: 13 ou une séquence d'acides aminés qui est identique à 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, ou 99% à la séquence d'acides aminés SEQ ID NO: 13.


 
6. Anticorps humanisé ou un fragment de celui-ci selon l'une quelconque des revendications précédentes, lequel anticorps est l'isotype IgG4.
 
7. Anticorps humanisé ou un fragment de celui-ci selon l'une quelconque des revendications précédentes, dans lequel ledit anticorps humanisé ou un fragment de celui-ci est capable d'inhiber l'agrégation des monomères de β-amyloïde aux fibrilles polymères de poids moléculaire élevé et de désagréger les fibrilles polymères préformées ou les filaments.
 
8. Anticorps humanisé ou un fragment de celui-ci selon l'une quelconque des revendications précédentes, dans lequel ledit anticorps humanisé ou un fragment de celui-ci se lie à une fibre Aβ, une fibrille ou un filament avec une affinité de liaison qui est d'au moins 10 fois, d'au moins 15 fois, d'au moins 20 fois, ou d'au moins 25 fois supérieures à l'affinité de liaison dudit anticorps ou d'un fragment de celui-ci à un monomère Aβ.
 
9. Anticorps humanisé ou un fragment de celui-ci selon l'une quelconque des revendications précédentes, dans lequel

(a) l'acide aminé en position 47 selon Kabat des régions charpentes de la région variable de la chaîne lourde est l'acide aminé Leu; et / ou

(b) l'acide aminé en position 94 selon Kabat dans les régions charpentes de la région variable de la chaîne lourde est l'acide aminé Ser; et / ou

(c) l'acide aminé en position 87 selon Kabat dans les régions charpentes de la région variable de la chaîne légère est l'acide aminé Tyr, Phe, Leu, VaI, Ile ou Ala.


 
10. Molécule d'acide nucléique comprenant une séquence nucléotidique codant pour l'anticorps humanisé ou un fragment de celui-ci selon l'une quelconque des revendications 1 à 9.
 
11. Vecteur d'expression comprenant la séquence nucléotidique de la revendication 10.
 
12. Cellule comprenant un vecteur d'expression selon la revendication 11.
 
13. Composition comprenant l'anticorps humanisé ou un fragment de celui-ci selon l'une quelconque des revendications 1 à 9, comprenant en outre un support pharmaceutiquement acceptable, un diluant et / ou un excipient.
 
14. Mélange comprenant l'anticorps humanisé ou un fragment de celui-ci selon l'une quelconque des revendications 1 à 9, comprenant en outre une substance biologiquement active, un support pharmaceutiquement acceptable, un diluant et / ou un excipient.
 
15. Mélange selon la revendication 14, comprenant

(i) une substance biologiquement active qui est un composé utilisé dans le traitement de l'amyloïdose, ou

(ii) au moins un des composés suivants: un composé contre le stress oxydatif; un composé anti-apoptotique; un chélateur de métaux; un inhibiteur de réparation d'ADN tel que la pirenzépine et ses métabolites; l'acide 3-amino-1-propanesulfonique (3 APS); le 1,3-propanedisulfonate (1,3PDS); un activateur de l'α-sécrétase; un inhibiteur de l'β-sécrétase; un inhibiteur de la γ-sécrétase; une protéine tau; un neurotransmetteur; un composé de rupture de feuillet β; un attractant pour la suppression / l'appauvrissement en amyloïde des composants cellulaires; un inhibiteur de la bêta amyloïde tronquée en position N-terminale, telle que l'amyloïde pyroglutamate bêta 3-42; une molécule anti-inflammatoire; un inhibiteur de la cholinestérase (ChEI) tel que la tacrine, la rivastigmine, le donepezil et / ou la galantamine; un agoniste M1; ou un autre médicament tel qu'un médicament modificateur de l'amyloïde ou de tau ou un complément nutritif.


 
16. L'anticorps ou un fragment de celui-ci selon l'une quelconque des revendications 1 à 9 , une composition selon la revendication 13, ou un mélange selon la revendication 14 ou 15 pour une utilisation dans une méthode de traitement ou d'atténuation des effets de l'amyloïdose chez un animal, tel qu'un mammifère ou un humain.
 
17. Anticorps humanisé ou un fragment de celui-ci, le mélange, ou la composition pour une utilisation selon la revendication 16, dans laquelle l'amyloïdose est l'amyloïdose secondaire, l'amyloïdose liée à l'âge, un trouble neurologique, la maladie d'Alzheimer (AD), la démence du corps de Lewy, le syndrome de Down, une hémorragie cérébrale héréditaire avec une amyloïdose (type néerlandais), le complexe de démence parkinsonienne de Guam, la paralysie supranucléaire progressive, la sclérose en plaques, la maladie de Creutzfeld Jacob, la maladie de Parkinson, la démence liée au VIH, la SLA (sclérose latérale amyotrophique), le diabète adulte, l'amyloïdose cardiaque sénile, les tumeurs endocrines ou dégénérescence maculaire.
 
18. Anticorps humanisé ou un fragment de celui-ci, le mélange ou la composition pour une utilisation selon la revendication 17, dans laquelle l'amyloïdose est la maladie d'Alzheimer.
 
19. Anticorps humanisé ou un fragment de celui-ci, le mélange, ou la composition pour une utilisation selon la revendication 16, dans laquelle le traitement de l'animal conduit à

i) une augmentation de la capacité de mémoire cognitive et / ou;

ii) la rétention de la capacité de mémoire cognitive; et / ou

iii) une restauration complète de la capacité de mémoire cognitive.


 
20. Procédé de diagnostic d'une amyloïdose chez un patient comprenant:

i) une mise en contact d'un échantillon, d'une partie du corps ou d'une zone corporelle contenant une protéine amyloïde avec l'anticorps humanisé ou un fragment de celui-ci selon l'une quelconque des revendications 1 à 9 dans des conditions permettant la liaison dudit anticorps ou un fragment de celui-ci contre la protéine amyloïde; et

ii) la détection de l'anticorps ou un fragment de celui-ci lié à la protéine,
de sorte que la présence ou l'absence d'anticorps ou un fragment de celui-ci lié à la protéine amyloïde indique la présence ou l'absence de protéine amyloïde dans l'échantillon, la partie du corps ou la zone corporelle.


 
21. Procédé de détermination de l'étendue du fardeau de plaque amyloïdogène dans un échantillon de tissu ou un échantillon de fluide corporel comprenant:

i) tester d'un échantillon de tissu ou d'un échantillon de fluide corporel pour la présence de protéine amyloïde avec l'anticorps humanisé ou un fragment de celui-ci selon l'une quelconque des revendications 1-9;

ii) déterminer la quantité d'anticorps ou un fragment de celui-ci liée à la protéine amyloïde; et

iii) calculer le fardeau de plaque dans l'échantillon de tissu ou l'échantillon de fluide corporel.


 
22. Kit pour la détection et le diagnostic de maladies et de conditions associées à l'amyloïde comprenant, l'anticorps humanisé ou un fragment de celui-ci selon l'une quelconque des revendications 1 à 9, dans lequel ledit kit comprenant un récipient contenant un ou plusieurs anticorps ou fragments de ceux-ci selon l'une quelconque des revendications 1 à 9 set des instructions pour l'utilisation desdits anticorps ou fragments de ceux-ci.
 
23. Anticorps humanisé ou un fragment de celui-ci selon l'une quelconque des revendications 1 à 9, une composition selon la revendication 13, ou un mélange selon la revendication 14 ou 15, pour une utilisation dans une méthode de prévention de la dégénérescence des neurones lors de l'exposition à des oligomères β-amyloïde, ou pour une utilisation dans une méthode de protection des neurones contre une dégradation induite par la protéine β-amyloïde.
 
24. Procédé pour une préparation d'un anticorps humanisé ou d'un fragment de celui-ci selon l'une quelconque des revendications 1 à 9, dans lequel le procédé comprend la culture de la cellule de la revendication 12 et la purification de l'anticorps ou d'un fragment de celui-ci.
 




Drawing










































































Cited references

REFERENCES CITED IN THE DESCRIPTION



This list of references cited by the applicant is for the reader's convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.

Patent documents cited in the description




Non-patent literature cited in the description